WO2017201501A1 - Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers - Google Patents

Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers Download PDF

Info

Publication number
WO2017201501A1
WO2017201501A1 PCT/US2017/033688 US2017033688W WO2017201501A1 WO 2017201501 A1 WO2017201501 A1 WO 2017201501A1 US 2017033688 W US2017033688 W US 2017033688W WO 2017201501 A1 WO2017201501 A1 WO 2017201501A1
Authority
WO
WIPO (PCT)
Prior art keywords
riluzole
cancer
agent
day
glutamate
Prior art date
Application number
PCT/US2017/033688
Other languages
French (fr)
Inventor
Vladimir Coric
Original Assignee
Biohaven Pharmaceutical Holding Company Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to LTEPPCT/US2017/033688T priority Critical patent/LT3458053T/en
Priority to DK17800310.9T priority patent/DK3458053T3/en
Priority to EP21207246.6A priority patent/EP4019019B1/en
Priority to US16/302,284 priority patent/US11400155B2/en
Application filed by Biohaven Pharmaceutical Holding Company Ltd. filed Critical Biohaven Pharmaceutical Holding Company Ltd.
Priority to JP2018560520A priority patent/JP7169195B2/en
Priority to RS20220165A priority patent/RS62935B1/en
Priority to PL17800310T priority patent/PL3458053T3/en
Priority to SI201731061T priority patent/SI3458053T1/en
Priority to EP17800310.9A priority patent/EP3458053B1/en
Priority to ES17800310T priority patent/ES2905823T3/en
Priority to HRP20220237TT priority patent/HRP20220237T1/en
Publication of WO2017201501A1 publication Critical patent/WO2017201501A1/en
Priority to CY20221100131T priority patent/CY1124999T1/en
Priority to US17/878,121 priority patent/US20230310595A1/en
Priority to JP2022173334A priority patent/JP2023012508A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/428Thiazoles condensed with carbocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39566Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against immunoglobulins, e.g. anti-idiotypic antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • C07D277/68Benzothiazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D277/82Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered

Definitions

  • the present invention relates to the use of riluzole, analogs of riluzole, prodrugs of riluzole and other related riluzole compounds to enhance the therapeutic effects of immunotherapeutic agents to treat oncologic diseases or cancers.
  • Riluzole (6-(trifluoromethoxy)benzothiazol-2-amine) is a pharmaceutical which has been used for treatment of amyotrophic lateral sclerosis (ALS). Recently, riluzole has been shown to have other clinical benefits. For example, orally administered riluzole dosed twice a day at a total dose of 100 mg may relieve or treat neuropsychiatric symptoms and disorders, such as mood, anxiety disorder, refractory depression, obsessive-compulsive anxiety and the like. Similarly, there is some indication that high doses of riluzole may have some anti-cancer effects but it has not yet been demonstrated to possess therapeutic anti-cancer effects on its own.
  • ALS amyotrophic lateral sclerosis
  • immuno-oncology targets include: CTLA4, cytotoxic T-lymphocyte-associated antigen 4; Ig, immunoglobulin; LAG3, lymphocyte activation gene 3; mAbs, monoclonal antibodies; PD-1, programmed cell death protein 1; PDL, PD-1 ligand; TIM3, T cell membrane protein 3, CD40L, A2aR, adenosine A2a receptor; B7RP1, B7-related protein 1; BTLA, B and T lymphocyte attenuator; GAL9, galectin 9;
  • HVEM herpesvirus entry mediator
  • ICOS inducible T cell co-stimulator
  • IL interleukin
  • KIR killer cell immunoglobulin-like receptor
  • PD-1 programmed cell death protein 1
  • PDL PD-1 ligand
  • TGF transforming growth factor- ⁇
  • TIM3 T cell membrane protein 3
  • CD27 CD27
  • immune targets include: Anti-VEGF-2 monoclonal antibody (Mab), Anti-EGFr Mab, IDOl inhibitor, Anti-B7-H3 Mab, Anti-GITR Mab, Anti-CD137 Mab, Anti-CD20 Mab, IL-15 superagonist/IL-15Ra-Fc fusion protein, Anti-CXCR4 Mab, Interleukin- 21, Interleukin-21, Anti-KIR Mab, Anti-CD27 Mab, Anti-CSF-IR Mab, Anti-CTLA- 4 MAb + GMCSF, Anti-CD30 MAb, Anti-LAG3 Mab, Anti-CD 19 Mab, Anti-OX40 Mab, Anti-CD73 Mab, OX40 agonist, or other agents including bi-specific molecules, small molecules targeting the immune system or anti-drug conjugates or vaccines.
  • Mab Anti-VEGF-2 monoclonal antibody
  • Anti-EGFr Mab IDOl inhibitor
  • Anti-B7-H3 Mab Anti-GITR Mab
  • Programed Death 1 (PD-1), an inhibitory checkpoint molecule, is expressed on T cells to limit peripheral immune responses.
  • Ligation of PD-1 with its corresponding ligands B7-H1 (PD-L1) or B7-DC (PD-L2) has been shown to result in direct inhibition of T-cell effector activation and T cell "exhaustion.”
  • PD-L1 and PD-L2 have been shown to be up-regulated on tumor cells in a variety of human cancers, representing a potential mechanism of immune escape.
  • expression of PD-1 is increased in tumor-infiltrating lymphocytes. Anti-PD-1 can block the increase in PD-1 or modify its effects.
  • Riluzole has multiple modes of action, including acting as a glutamate modulating agent.
  • Glutamine a "conditionally" essential amino acid has been demonstrated to be paramount to macromolecular synthesis and tumor cell metabolism.
  • a variety of solid malignancies have been shown to overexpress phosphate-dependent glutaminase (GLS), which converts glutamine to glutamate further emphasizing the role of glutamine in cancer metabolism.
  • GLS phosphate-dependent glutaminase
  • glutamate is a key nitrogen "waste" bank and critical in a variety of cellular metabolic pathways. As such, reduction in glutamine/glutamate levels to immune cells may reduce proliferative and effector function, limiting an anti-tumor immune mediated response.
  • Glutamate modulators such as riluzole may be effective as part of a combination therapy with anti-cancer agents targeting the immune system to treat disease.
  • glutamate modulators such as riluzole are used with immunotherapeutic agents, such as certain anti-cancer agents, to treat proliferative diseases such as cancer. Analogs of riluzole may also have similar effects.
  • glutamate modulating agents include but are not limited to memantine, n-acetlcysteine, amantadine, topiramate, pregabalin, lamotrigine, ketamine, s-ketamine, AZD8108, AZD 6765, BHV-4157,
  • glutamate modulating agents also include but are not limited to NMDA receptor antagonists, kainite receptor antagonists, AMPA receptor anatagonists, metabotropic glutamate receptors, or agents that target excitatory amino acide transporters or vescular glutamate transport.
  • glutamate modulators may cause a reduction in the glutamine/glutamate levels or increase the cycling of glutamate by increasing the expression of excitatory amino acid transporters, causing a reduction in reduce proliferative and effector function.
  • a combination therapy employing riluzole (or related compounds) with an immunotherapy agent or other anti-cancer drug, provided simultaneously or sequentially, may have excellent cancer treating properties. In fact, there may be synergistic effects. Accordingly, the present invention relates to such a combination.
  • the present invention uses a combination immunotherapy having a glutamate modulating agent and an immunotherapy agent to treat disease, particularly cancer.
  • a preferred glutamate modulating agent is riluzole and a preferred immunotherapy agent is a checkpoint inhibitor such as an anti-PD-1. It appears that the glutamate modulators make the cancer cells more susceptible to the anti-cancer agents such as immunotherapeutic agents.
  • the glutamate modulating agents may be given orally, sublingually, subcutaneously or in any other means of delivery.
  • the glutamate modulating agents may be in the form of a prodrug, which releases the agent in the body, a sustained release vehicle, a delayed release vehicle, or any other delivery form.
  • the glutamate modulating agent and the immunotherapy agent may be delivered simultaneously or sequentially. If the agents are delivered sequentially, either agent may be dosed first, and the separation of time may include finishing the dosing of one agent completely before commencing the dosage of the other or they may be intermingled in time.
  • the preferred glutamate modulating agents include but are not limited to: amantadine, lamotrigine, memantine, with riluzole and its prodrugs being most preferred. Prodrugs of riluzole are described in United States Patent Application Serial No. 14/385,551, United States Patent Application Serial No. 14/410,647, PCT Application Serial No. PCT/US2016/019773 and PCT Application Serial
  • a sublingual formulation useful in the present invention comprises an effective amount of riluzole or pharmaceutically acceptable salts, solvates, anomers, enantiomers, hydrates or prodrugs thereof.
  • the formulation provides sufficient solubility for riluzole to be incorporated into the sublingual formulation at relatively large doses and sublingually delivered.
  • the formulation is preferably a modified oral disintegrating formulation of riluzole.
  • the excipients, including mannitol and gelatin are blended, solubilized with water and deaerated before being mixed with the active pharmaceutical ingredient (or "API"), riluzole, which has been milled separately.
  • API active pharmaceutical ingredient
  • Particle size of the API (D50) is less than about 2 microns.
  • the mixture is lyophilized by flash freezing and then freeze-dried.
  • the formulation has good oral palatability.
  • the effective amount of glutamate modulating agent for the sublingual formulation useful in the present invention to achieve a lower therapeutic dose may be less than that of orally administered agent.
  • effective dose of the sublingual formulation of the glutamate modulating agent may be about 1 to 95 % of that of the orally administered agent.
  • a sublingual formulation of the immunotherapeutic agent it may also have improved properties.
  • the glutamate modulating agent as part of the formulation for treating cancer or symptoms may be dosed at or below about 400 mg/day, at or below about 300 mg/day, at or below about 150 mg/day, at or below about 100 mg/day, at or below about 70 mg/day, at or below about 60 mg/day, at or below about 50 mg/day, at or below about 42.5 mg/day, at or below about 37.5 mg/day at or below about 35 mg/day, at or below about 20 mg/day, at or below about 17.5 mg/day, at or below about 15 mg/day, at or below about 10 mg/day, at or below about 5 mg/day, or at or below about 1 mg/day.
  • the immunotherapeutic agent should be dosed at about 1-100 mg/kg; for example, 1 mg/kg, 2 mg, kg, 5 mg/kg, 7.5 mg/kg, 10 mg/kg, 20 mg/kg, 25 mg/kg, 50 mg/kg, 75 mg/kg, 100 mg/kg, or any intermediate values.
  • a preferred immunotherapeutic agent is anti-PD-1. Dosing may be daily, alternate days, weekly or having an even higher time separation. In certain circumstances, more often dosing can be used.
  • the glutamate modulating agent and immunotherapeutic agent can be delivered simultaneously or sequentially.
  • riluzole refers to a drug having a chemical structure as follows. It is currently available in the market as RILUTEK®.
  • riluzole also refers to all prodrugs, enantiomers, or derivatives and its
  • sublingual administration refers to a route of administrating a chemical agent or a drug by placing thereof under a tongue of a subject.
  • prodrug as used herein, is a precursor of a drug which may be administered in an altered or less active form.
  • the prodrug may be converted into the active drug form in physiological environments by hydrolysis or other metabolic pathways.
  • riluzole prodrug refers to a compound which is a derivative from riluzole with modification therein.
  • a riluzole prodrug may also refer to a compound that is metabolized into an active form of riluzole by the body.
  • ALS Amyotrophic Lateral Sclerosis
  • immunotherapeutic anti-cancer agent includes any agent that targets the immune system to result in an anti-cancer therapeutic effects.
  • targets and agents include but are not limited to: anti-PD-1, anti-PD-Ll, anti-CTLA4 or other immunotherapy or checkpoint inhibitor targets.
  • immuno-oncology targets include: CTLA4, cytotoxic T-lymphocyte-associated antigen 4; Ig, immunoglobulin; LAG3, lymphocyte activation gene 3; mAbs, monoclonal antibodies; PD-1, programmed cell death protein 1; PDL, PD-1 ligand; TIM3, T cell membrane protein 3, CD40L, A2aR, adenosine A2a receptor; B7RP1, B7-related protein 1; BTLA, B and T lymphocyte attenuator; GAL9, galectin 9; HVEM, herpesvirus entry mediator; ICOS, inducible T cell co-stimulator; IL, interleukin; KIR, killer cell immunoglobulin-like receptor; LAG3, lymphocyte activation gene 3; PD-1, programmed cell death protein 1; PDL, PD-1 ligand; TGF , transforming growth factor- ⁇ ; TIM3, T cell membrane protein 3; and CD27.
  • CTLA4 cytotoxic T-lymphocyte-associated
  • immune targets include: Anti-VEGF-2 monoclonal antibody (Mab), Anti-EGFr Mab, IDOl inhibitor, Anti-B7-H3 Mab, Anti-GITR Mab, Anti-CD 137 Mab, Anti-CD20 Mab, IL- 15 superagonist/IL-15Ra-Fc fusion protein, Anti-CXCR4 Mab, Interleukin-21, Interleukin-21, Anti-KIR Mab, Anti-CD27 Mab, Anti-CSF-IR Mab, Anti-CTLA-4 MAb + GMCSF, Anti-CD30 MAb, Anti-LAG3 Mab, Anti-CD19 Mab, Anti-OX40 Mab, Anti-CD73 Mab, OX40 agonist, or other agents including bi-specific molecules, small molecules targeting the immune system or anti-drug conjugates or vaccines, or nivolumab (Opdivo), pembrolizumab (Keytruda), pidilizumab, ipilimumab (Yervoy),
  • cancer includes, but is not limited to, the following proliferative diseases: Acute Lymphoblastic Leukemia (ALL), Acute Myeloid Leukemia (AML), Adrenocortical Carcinoms, Childhood cancers, AIDS-Related Cancers, Kaposi Sarcoma, AIDS-Related Lymphoma, Primary CNS Lymphoma, Anal Cancer, Astrocytomas, Atypical Teratoid/Rhabdoid Tumor, Basal Cell Carcinoma, Skin Cancer (Nonmelanoma), Bile Duct Cancer, Bladder Cancer, Bone Cancer, Ewing Sarcoma Family of Tumors, Osteosarcoma and Malignant Fibrous Histiocytoma, Brain Stem Glioma, Atypical Teratoid/Rhabdoid Tumor, Embryonal Tumors, Germ Cell Tumors, Craniopharyngioma, Ependymoma, Breast Cancer, Bronchial Tumors, Burkit
  • Trophoblastic Disease Glioma, Hairy Cell Leukemia, Head and Neck Cancer, Hepatocellular (Liver) Cancer, Histiocytosis, Langerhans Cell, Hodgkin Lymphoma, Hypopharyngeal Cancer, Islet Cell Tumors, Pancreatic Neuroendocrine Tumors, Kaposi Sarcoma, Kidney, Renal Cell, Langerhans Cell Histiocytosis, Laryngeal Cancer, Leukemia, Acute Lymphoblastic (ALL), Acute Myeloid (AML), Chronic Lymphocytic (CLL), Chronic Myelogenous (CML), Hairy Cell, Lip and Oral Cavity Cancer, Liver Cancer (Primary), Lung Cancer, Non-Small Cell, Small Cell, Lymphoma, Hodgkin, Non-Hodgkin, Macroglobulinemia, Waldenstrom, Male Breast Cancer, Melanoma, Merkel Cell Carcinoma, Mesothelioma,Metastatic Squamous Neck Cancer
  • Myelodysplastic/Myeloproliferative Neoplasms Myelogenous Leukemia, Chronic (CML), Myeloid Leukemia, Acute (AML) Myeloma, Multiple, Myeloproliferative Neoplasms, Nasal Cavity and Paranasal Sinus Cancer, Nasopharyngeal Cancer, Neuroblastoma, Non-Hodgkin Lymphoma, Non-Small Cell Lung Cancer, Oral Cancer, Oral Cavity Cancer, Lip and Oropharyngeal Cancer, Osteosarcoma and
  • Lymphoma Testicular Cancer, Throat Cancer, Thymoma and Thymic Carcinoma, Thyroid Cancer, Transitional Cell Cancer of the Renal Pelvis and Ureter, Unknown Primary, Ureter and Renal Pelvis, Transitional Cell Cancer, Urethral Cancer, Uterine Cancer, Endometrial, Uterine Sarcoma, Vaginal Cancer, Vulvar Cancer, Waldenstrom Macroglobulinemia, and Wilms Tumor.
  • treatment includes any treatment of a condition or disease in a subject, or particularly a human, and may include: (i) preventing the disease or condition from occurring in the subject which may be predisposed to the disease but has not yet been diagnosed as having it; (ii) inhibiting the disease or condition, i.e., arresting its development; relieving the disease or condition, i.e., causing regression of the condition; or (iii) ameliorating or relieving the conditions caused by the disease, i.e., symptoms of the disease.
  • Treatment could be used in combination with other standard therapies or alone.
  • the term “effective” is used to describe an amount of a compound, composition or component which, when used within the context of its intended use, effects an intended result.
  • an effective amount refers to that amount which is sufficient to effect treatment, as defined herein, when administered to a subject in need of such treatment.
  • the effective amount will vary depending on the subject and disease state being treated, the severity of the affliction and the manner of administration, and may be determined routinely by one of ordinary skill in the art.
  • pharmaceutically acceptable salt is used throughout the specification to describe, where applicable, a salt form of one or more of the compounds or prodrugs described herein which are presented to increase the solubility of the compound in the gastric or gastroenteric juices of the patient's gastrointestinal tract in order to promote dissolution and the bioavailability of the compounds.
  • Pharmaceutically acceptable salts include those derived from
  • Suitable salts include those derived from alkali metals such as potassium and sodium, alkaline earth metals such as calcium, magnesium and ammonium salts, among numerous other acids and bases well known in the pharmaceutical art.
  • Sodium and potassium salts are particularly preferred as neutralization salts of the phosphates according to the present invention.
  • the description provides pharmaceutically acceptable salts of the modified peptides as described herein, which retain the biological effectiveness and properties of the parent compounds and which are not biologically or otherwise harmful as the dosage administered.
  • the compounds of this invention are capable of forming both acid and base salts by virtue of the presence of amino and carboxy groups respectively.
  • Cmax refers to a maximum concentration of a drug in blood, serum, a specified compartment or test area of a subject between administration of a first dose and administration of a second dose.
  • Cmax could also refer to dose normalized ratios if specified.
  • Tmax refers to a time or period after
  • AUC area under the curve
  • AUC refers to a total amount of drug absorbed or exposed to a subject. Generally, AUC may be obtained from mathematical method in a plot of drug concentration in the subject over time until the concentration is negligible.
  • AUC area under the curve
  • the invention relates to a combination therapy utilizing an immunotherapeutic agent and a glutamate modulating agent.
  • the combination of these two drugs may be administered in a single dose as combined product, administered simultaneously using the same or distinct formats, or administered sequentially using the same or different forms of delivery.
  • the immunotherapeutic agent and the glutamate modulating agent can both be made into a tablet or part of a sublingual form, they can be administered together.
  • the immunotherapeutic agent can only be administered by injection (bolus or intravenous), and the glutamate modulating agent can be administered in the same format, this could also be used for simultaneous or sequential administration.
  • the immunotherapeutic agent can only be delivered by injection (for example, if it is an antibody), and the glutamate modulating agent can be delivered as a tablet or sublingually, delivery of the two agents can take place by differing formats.
  • PCT/US2015/061114 describe a sublingual formulation of riluzole, a preferred glutamate modulating agent.
  • the sublingual formulation may be administered in an effective amount to a subject in need thereof.
  • the subject may be an animal or human.
  • the glutamate modulating agent or its pharmaceutically acceptable salts thereof may be formulated in a pharmaceutical composition suitable for sublingual administration.
  • the immunotherapeutic agent may also be formulated as a sublingual, although injection is more standard.
  • Glutamate modulating agents such as riluzole and the pharmaceutically acceptable salts thereof can be formulated using pharmaceutically acceptable carriers well known in the art into dosages suitable for sublingual or buccal administration.
  • Such carriers enable the glutamate modulating agent for sublingual administration to be formulated in dosage forms such as tablets, powders, pills, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for sublingual absorption by a subject to be treated.
  • These carriers may be, but not limited to, selected from sugars, starches, cellulose and its derivatives, malt, gelatin, talc, calcium sulphate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffered solutions, emulsifiers, isotonic saline, pyrogen-free water and combinations thereof.
  • any form of substance may be accepted to sublingual administration if it dissolves easily in saliva.
  • the sublingually administered chemical agent or the drug can diffuse into capillaries through mucous membrane under the tongue, and then enter venous circulation of the subject.
  • sublingual administration may have advantages over oral administration as allowing for direct or faster entry to venous circulation, without risks of degradation in gastrointestinal tract, alteration by drug metabolism in liver and the like.
  • Various drugs in the market are designed for sublingual administration.
  • Riluzole is generally used to treat amyotrophic lateral sclerosis (ALS).
  • riluzole or prodrugs of riluzole or pharmaceutically acceptable salts thereof is subjected to a sublingual administration for the treatment of other disorders, including cancer.
  • the pharmaceutical composition may include an approved pharmaceutical ingredient, i.e., riluzole, in an effective amount to achieve their intended purpose.
  • an approved pharmaceutical ingredient i.e., riluzole
  • the dose of the glutamate modulating agent administered sublingually to the subject should be sufficient to provide a beneficial response in the subject over time such as reduction in symptoms in conjunction with the immunotherapeutic agent.
  • the combination may have synergistic effects.
  • the quantity of the glutamate modulating agent and the quantity of the immunotherapeutic agent to be administered may depend on the subject to be treated inclusive of the age, sex, weight and general health condition thereof. In this regard, precise amounts of the agent(s) for administration will depend on the judgment of the practitioner. In determining the effective amount of the glutamate modulating agent and immunotherapeutic agent to be administered in the treatment or reducing of the conditions associated with the symptoms and disorders, the physician may evaluate clinical factors including symptoms severity or progression of the disorder. In some conditions, a rapid absorption of the glutamate modulating agent or
  • immunotherapeutic agent may be desirable.
  • those of skill in the art may readily determine suitable dosages of the chemical agents of the invention.
  • the pharmaceutical composition also includes other pharmaceutically acceptable carriers and/or excipients such as binders, lubricants, diluents, coatings, disintegrants, barrier layer components, glidants, coloring agents, solubility enhancers, gelling agents, fillers, proteins, co-factors, emulsifiers, solubilizing agents, suspending agents and mixtures thereof.
  • binders such as binders, lubricants, diluents, coatings, disintegrants, barrier layer components, glidants, coloring agents, solubility enhancers, gelling agents, fillers, proteins, co-factors, emulsifiers, solubilizing agents, suspending agents and mixtures thereof.
  • binders such as binders, lubricants, diluents, coatings, disintegrants, barrier layer components, glidants, coloring agents, solubility enhancers, gelling agents, fillers, proteins, co-factors, emulsifiers, solubilizing
  • the pharmaceutical composition for sublingual use can be obtained by combining an approved pharmaceutical ingredient, i.e., riluzole, with further excipients, with optionally processing to obtain dosage forms such as tablets, powders, pills, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for sublingual absorption by a subject to be treated.
  • an approved pharmaceutical ingredient i.e., riluzole
  • dosage forms such as tablets, powders, pills, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for sublingual absorption by a subject to be treated.
  • Suitable excipients may be, but not limited to, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropyl methyl-cellulose, sodium carboxymethylcellulose, and/or polyvinyl-pyrrolidone (PVP).
  • fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol
  • cellulose preparations such as maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropyl methyl-cellulose, sodium carboxymethylcellulose, and/or polyvinyl-pyrrolidone (PVP).
  • PVP polyvinyl-pyrrolidone
  • disintegrating agents may be combined as well, and exemplary disintegrating agents may be, but not limited to, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • the compositions may be prepared by any of the methods of pharmacy but all methods include the step of bringing into association one or more chemical agents as described above with the carrier which constitutes one or more necessary ingredients.
  • the pharmaceutical compositions of the present invention may be manufactured in conventional methods known in the art, for example, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, lyophilizing processes and the like.
  • the sublingual formulation useful in the combination product of the invention may be prepared in a form of an orally dissolving or disintegrating tablet (ODT).
  • ODT as used herein may be prepared by mixing the glutamate modulating agent and/or the immunotherapeutic agent with water-soluble diluents and compressed in a tablet.
  • a suspension comprising the active product may be prepared with appropriate excipients and the suspension may be dispensed into blister packs and freeze-dried.
  • An exemplary freeze-dried preparation platform that could be used for the ODT is the ZYDIS® (Catalent, Somerset, NJ, USA) formulation.
  • the excipients, including water are blended and the glutamate modulating agent is separately milled to size and mixed with the excipients.
  • the sublingual formulation useful in the invention may comprise the glutamate modulating agent or an effective amount of a glutamate modulating agent prodrug.
  • the immunotherapeutic agent may be made into a prodrug.
  • the prodrug may be similar or less active form of the active.
  • the prodrug may have improved physiochemical, physiological pharmacokinetic or therapeutical characteristics when administered sublingually.
  • the prodrug may reduce side effects when orally or sublingually administered.
  • the clinical or therapeutic effect of the compound, or a subportion of the final product, sublingually formulated may have an improved pharmacokinetic profile for the pharmaceutical agent as measured by standard testing parameters.
  • the Tmax, Cmax and AUC of the drug may be improved compared to the same dose of the orally administered version of the same compound.
  • the sublingual formulation of the glutamate modulating agent may have a greater Cmax than the orally administered glutamate modulating agent to provide a therapeutically beneficial effect.
  • the sublingual formulation of the glutamate modulating agent may have an earlier or lesser Tmax than the orally administered glutamate modulating agent to provide a therapeutically beneficial effect and in some instances, a more rapid therapeutic effect.
  • the sublingual formulation of the glutamate modulating agent may have a greater AUC per milligram of the agent than the orally administered glutamate modulating agent.
  • the glutamate modulating agent may make the immunotherapeutic agent more effective, lesser amounts of the immunotherapeutic agent may be needed to achieve the same results, with a lessening of the inherent side effects.
  • the invention provides a method of treating a disease such as cancer.
  • the method comprises administering sublingually an effective amount of glutamate modulating agent or pharmaceutically acceptable salts thereof and an anti-cancer agent, preferably an immunotherapeutic agent, or pharmaceutically acceptable salts thereof to a subject in need thereof.
  • Identifying the subject in need of such treatment can be in the judgment of the subject or a health care professional and can be subjective (e.g., opinion) or objective (e.g., measurable by a test or diagnostic method).
  • the identified subject may be an animal or human in need thereof, particularly a human.
  • Such treatment will be suitably administered to subjects, particularly humans, suffering from the disease.
  • the effective amount of the treatment will vary depending on the subject and disease state being treated, the severity of the affliction and the manner of
  • the therapeutic effect of the combination product may be evident to occur within about a few minutes to about an hour after administration thereof.
  • the therapeutic effect may begin within about 1 minute, within about 2 minutes, within about 3 minutes, within about 4 minutes, within about 5 minutes, within about 6 minutes, within about 7 minutes, within about 8 minutes, within about 9 minutes, within about 10 minutes, within about 11 minutes, within about 12 minutes, within about 13 minutes, within about 14 minutes, within about 15 minutes, within about 16 minutes, within about 17 minutes, within about 18 minutes, within about 20 minutes, within about 60 minutes, or within about 90 minutes after administration.
  • long term cure or amelioration of the disease may not occur for weeks or months after administration.
  • the effects on the symptoms may be maintained for about 1 hour, for about 2 hours, for about 3 hours, for about 4 hours, for about 5 hours, for about 6 hours m for about 7 hours, for about 8 hours, for about 9 hours, for about 10 hours, for about 12 hours, for about 14 hours, for about 16 hours, for about 18 hours, for about 20 hours, for about 22 hours, for about 24 hours, for about 2 days, or for about 3 days or more after administration thereof.
  • the effective amount or dose of glutamate modulating agent for sublingual administration may be less than that of orally administered agent.
  • the effective dose in sublingual administration of the glutamate modulating agent may be of about 1-95 % of the dose of the orally administered agent itself.
  • similar reduction in the amount of the immunotherapeutic agent may be achieved by administration of the glutamate modulating agent itself, by any mode of administration.
  • Optional dosage frequencies include once a day, twice a day, three times a day, four times a day, once every other day, once a week, twice a week, three times a week, four times a week, once every two weeks, once or twice monthly, and the like.
  • Glutamate modulating agents can be used as is or may be in the form of prodrugs.
  • Prodrugs of riluzole are described in United States Patent Application Serial No. 14/385,551, United States Patent Application Serial No. 14/410,647, PCT Application Serial No. PCT/US2016/019773 and PCT Application Serial
  • the preferred riluzole prodrugs have the structure:
  • P 23 is selected from the group consisting H, CH 3 , CH2CH3, CH2CH2CH3, CH2CCH, CH(CH 3 ) 2 , CH 2 CH(CH 3 ) 2 , CH(CH 3 )CH 2 CH 3 , CH2OH, CH 2 OCH 2 Ph,
  • a preferred prodrug of riluzole has the following formula:
  • prodrugs can be made from other glutamate modulating agents. Such agents are useful as part of the combination of the present invention.
  • GL261-Luc cells are grown in Dulbecco's Modified Eagle Medium (DMEM) + 10% fetal bovineserum + 1% penicillinstreptomycin at 37°C in a humidified incubator maintained at 5% CO and 5% O2 (Gibco).
  • mice Female C57BL/6J mice (Harlan), 4 to 6 weeks old or 6 to 8 weeks old, are used for orthotopic glioma experiments as described in Sonabend AM, Velicu S, Ulasov IV, et al. A safety and efficacy study of local delivery of interleukinl2 transgene by PPC polymer in a model of experimental glioma. Anticancer Drugs. 2008;19: 133-142.
  • To establish syngeneic gliomas 130,000 GL261-Luc cells are stereotactically injected in a ⁇ volume into the left striatum over 1 minute into the following coordinates: 1 mm anterior, 1 mm lateral from bregma, and 3 mm deep from the cortical surface.
  • Tumor burden is monitored by luciferase imaging on days 7, 21 and 35 after implantation, and the mice are randomly allocated into treatment arms based on tumor radiance, so that the average tumor radiance in each group is roughly equivalent.
  • the animals are euthanized when they show predetermined signs of neurologic deficits (failure to ambulate, weight loss >20%body mass, lethargy, hunched posture).
  • the tumor take rate is 100%.
  • Each arm has 6 to 10 mice in survival experiments. All experiments are repeated at least in triplicate.
  • Hamster antimurine PD-1 monoclonal antibody producing hybridoma G4 are used to produce antibodies as described in Hirano F, Kaneko K, Tamura H, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005;65: 1089-1096.
  • mice Female C57BL/6J mice, 4 to 6 weeks old, were implanted intrancranially in the left striatum with 130,000 GL261 cells each. The mice were housed and maintained according to the institutional Animal Care and Use Committee protocol in the Johns Hopkins University Animal Facility. The mice were imaged by
  • Day 0 represents the date of intracranial implantation.
  • Control arm 1 received no treatment.
  • Control arm 2 received aPD-1 alone at a dose of 200 ug/animal via intraperitoneal injection on days 10, 12, 14.
  • Control arms 3, 4 and 5 received BHV- 4157 alone at doses of 15, 30 and 45 mg/kg (respectively) via intraperitoneal injection daily beginning on day 10.
  • Control arms 6, 7 and 8 received BHV-4157 at doses of 15, 30 and 45 mg/kg (respectively) via intraperitoneal injection daily beginning on day 10 and aPD-1 at a dose of 200 ug/animal via intraperitoneal injection on days 10, 12, 14.
  • MSRx refers to a value calculated by dividing: (i) the percentage survival of mice treated with an immunotherapeutic anti-cancer agent plus a glutamate modulating agent, by (ii) the percentage survival of mice treated with an immunotherapeutic anti-cancer agent alone, in accordance with the procedure set forth in Example 1 hereof at a time period of "x" number of days after implantation of the tumor into the mice.
  • MSR60 refers to the Mouse Survival Ratio at a time of 60 days after tumor implantation.
  • mice in Arm 1 had 0% survival
  • the mice in Arm 2 had 50% survival
  • the mice in Arms 6, 7 and 8 had at least 70 to 80% survival.
  • the Mouse Survival Ratio was about 1.4 to 1.6 (i.e., 70/50 and 80/50).
  • the mice in Arm 1 had 0% survival
  • the mice in Arm 2 had 30% survival
  • the mice in Arms 6, 7 and 8 had at least 60 to 80% survival.
  • the Mouse Survival Ratio (MSR28) was about 2.0 to 2.6 (i.e., 60/30 and 80/30).
  • the Mouse Survival Ratio was about 2.0 to 2.3 (i.e., 60/30 and 70/30).
  • the Mouse Survival Ratio is at least 1.4, more preferably at least 1.6 when measured at 26 days after tumor implantation (MSR26).
  • the Mouse Survival Ratio is at least 2.0, more preferably at least 2.6 when measured at 28 days after tumor implantation (MSR28).
  • the Mouse Survival Ratio is at least 2.0, more preferably at least 2.3 when measured at 60 days after tumor implantation (MSR60).
  • the Mouse Survival Ratio measured at a time when the untreated mice reach 0% survival, or thereafter until a time of 60 days after tumor implantation is at least 1.4, at least 1.6, at least 2.0, at least 2.3 or at least 2.6.
  • combination therapy i.e., an immunotherapeutic anti-cancer agent and a glutamate modulating agent
  • a Mouse Survival Ratio of at least 2.0, more typically at least 2.3 (measured at day 60, MSR60).
  • glutamate receptors are found on a number of other tumor cells and it is believed that this combination therapy could be effective for those cells as well.

Abstract

Disclosed are methods of treating cancer using a combination of an immunotherapeutic agent, such as, for example, a PD-1, PD-L1 or CTLA-4 checkpoint inhibitor, and a glutamate modulating agent such as riluzole or trigriluzole. Pharmaceutical compositions including the immunotherapeutic agents and glutamate modulating agents are also disclosed.

Description

USE OF RILUZOLE. RILUZOLE PRODRUGS OR RILUZOLE ANALOGS WITH IMMUNOTHERAPIES TO TREAT CANCERS
Cross Reference to Related Applications
This application claims the benefit of U.S. Provisional Application Serial Number 62/339,433 filed May 20, 2016.
Field of the Invention
The present invention relates to the use of riluzole, analogs of riluzole, prodrugs of riluzole and other related riluzole compounds to enhance the therapeutic effects of immunotherapeutic agents to treat oncologic diseases or cancers.
Background of the Invention
Riluzole (6-(trifluoromethoxy)benzothiazol-2-amine) is a pharmaceutical which has been used for treatment of amyotrophic lateral sclerosis (ALS). Recently, riluzole has been shown to have other clinical benefits. For example, orally administered riluzole dosed twice a day at a total dose of 100 mg may relieve or treat neuropsychiatric symptoms and disorders, such as mood, anxiety disorder, refractory depression, obsessive-compulsive anxiety and the like. Similarly, there is some indication that high doses of riluzole may have some anti-cancer effects but it has not yet been demonstrated to possess therapeutic anti-cancer effects on its own.
In recent years, a number of anti-cancer therapies targeting the immune system have demonstrated robust efficacy across many tumor types. Despite this novel approach to treat cancers with emerging immunotherapeutic agents, many patients do not show a complete response or remission to treatment with immunotherapies. Also, certain combination immuno-oncologic agents have significant toxicity (primarily auto-immune reactions). There is an urgent need to further augment or enhance the effect of immune-oncology therapies. Examples of therapeutic approaches to cancer with immunology targeting anti-cancer agents include: anti-PD-1, anti-PD-Ll, anti- CTLA4 or other immunotherapy or checkpoint inhibitor targets. Other examples of immuno-oncology targets include: CTLA4, cytotoxic T-lymphocyte-associated antigen 4; Ig, immunoglobulin; LAG3, lymphocyte activation gene 3; mAbs, monoclonal antibodies; PD-1, programmed cell death protein 1; PDL, PD-1 ligand; TIM3, T cell membrane protein 3, CD40L, A2aR, adenosine A2a receptor; B7RP1, B7-related protein 1; BTLA, B and T lymphocyte attenuator; GAL9, galectin 9;
HVEM, herpesvirus entry mediator; ICOS, inducible T cell co-stimulator; IL, interleukin; KIR, killer cell immunoglobulin-like receptor; LAG3, lymphocyte activation gene 3; PD-1, programmed cell death protein 1; PDL, PD-1 ligand; TGF , transforming growth factor-β; TIM3, T cell membrane protein 3; and CD27. Other immune targets include: Anti-VEGF-2 monoclonal antibody (Mab), Anti-EGFr Mab, IDOl inhibitor, Anti-B7-H3 Mab, Anti-GITR Mab, Anti-CD137 Mab, Anti-CD20 Mab, IL-15 superagonist/IL-15Ra-Fc fusion protein, Anti-CXCR4 Mab, Interleukin- 21, Interleukin-21, Anti-KIR Mab, Anti-CD27 Mab, Anti-CSF-IR Mab, Anti-CTLA- 4 MAb + GMCSF, Anti-CD30 MAb, Anti-LAG3 Mab, Anti-CD 19 Mab, Anti-OX40 Mab, Anti-CD73 Mab, OX40 agonist, or other agents including bi-specific molecules, small molecules targeting the immune system or anti-drug conjugates or vaccines.
Programed Death 1 (PD-1), an inhibitory checkpoint molecule, is expressed on T cells to limit peripheral immune responses. Ligation of PD-1 with its corresponding ligands B7-H1 (PD-L1) or B7-DC (PD-L2) has been shown to result in direct inhibition of T-cell effector activation and T cell "exhaustion." PD-L1 and PD-L2 have been shown to be up-regulated on tumor cells in a variety of human cancers, representing a potential mechanism of immune escape. Furthermore, expression of PD-1 is increased in tumor-infiltrating lymphocytes. Anti-PD-1 can block the increase in PD-1 or modify its effects.
Riluzole has multiple modes of action, including acting as a glutamate modulating agent. Glutamine, a "conditionally" essential amino acid has been demonstrated to be paramount to macromolecular synthesis and tumor cell metabolism. A variety of solid malignancies have been shown to overexpress phosphate-dependent glutaminase (GLS), which converts glutamine to glutamate further emphasizing the role of glutamine in cancer metabolism. However, glutamate is a key nitrogen "waste" bank and critical in a variety of cellular metabolic pathways. As such, reduction in glutamine/glutamate levels to immune cells may reduce proliferative and effector function, limiting an anti-tumor immune mediated response. Glutamate modulators such as riluzole may be effective as part of a combination therapy with anti-cancer agents targeting the immune system to treat disease. In particular, glutamate modulators such as riluzole are used with immunotherapeutic agents, such as certain anti-cancer agents, to treat proliferative diseases such as cancer. Analogs of riluzole may also have similar effects. A number of other glutamate modulating agents are known. These include but are not limited to memantine, n-acetlcysteine, amantadine, topiramate, pregabalin, lamotrigine, ketamine, s-ketamine, AZD8108, AZD 6765, BHV-4157,
dextromethorphan, AV-101, CERC-301, GLY-13, and prodrugs or analogs thereof. These glutamate modulating agents also include but are not limited to NMDA receptor antagonists, kainite receptor antagonists, AMPA receptor anatagonists, metabotropic glutamate receptors, or agents that target excitatory amino acide transporters or vescular glutamate transport. These glutamate modulators may cause a reduction in the glutamine/glutamate levels or increase the cycling of glutamate by increasing the expression of excitatory amino acid transporters, causing a reduction in reduce proliferative and effector function.
A combination therapy employing riluzole (or related compounds) with an immunotherapy agent or other anti-cancer drug, provided simultaneously or sequentially, may have excellent cancer treating properties. In fact, there may be synergistic effects. Accordingly, the present invention relates to such a combination.
Summary of the Invention
The present invention uses a combination immunotherapy having a glutamate modulating agent and an immunotherapy agent to treat disease, particularly cancer. A preferred glutamate modulating agent is riluzole and a preferred immunotherapy agent is a checkpoint inhibitor such as an anti-PD-1. It appears that the glutamate modulators make the cancer cells more susceptible to the anti-cancer agents such as immunotherapeutic agents.
The glutamate modulating agents may be given orally, sublingually, subcutaneously or in any other means of delivery. The glutamate modulating agents may be in the form of a prodrug, which releases the agent in the body, a sustained release vehicle, a delayed release vehicle, or any other delivery form. The glutamate modulating agent and the immunotherapy agent may be delivered simultaneously or sequentially. If the agents are delivered sequentially, either agent may be dosed first, and the separation of time may include finishing the dosing of one agent completely before commencing the dosage of the other or they may be intermingled in time.
The preferred glutamate modulating agents include but are not limited to: amantadine, lamotrigine, memantine, with riluzole and its prodrugs being most preferred. Prodrugs of riluzole are described in United States Patent Application Serial No. 14/385,551, United States Patent Application Serial No. 14/410,647, PCT Application Serial No. PCT/US2016/019773 and PCT Application Serial
No.PCT/US2016/019787, the disclosures of which are all incorporated herein by reference. Sublingual formulations of riluzole that provide stability and excellent properties are described in PCT Application Serial No. PCT/US2015/061106 and PCT Application Serial No. PCT/US2015/061114, the disclosures of which are also incorporated by reference.
A sublingual formulation useful in the present invention comprises an effective amount of riluzole or pharmaceutically acceptable salts, solvates, anomers, enantiomers, hydrates or prodrugs thereof. The formulation provides sufficient solubility for riluzole to be incorporated into the sublingual formulation at relatively large doses and sublingually delivered. The formulation is preferably a modified oral disintegrating formulation of riluzole. The excipients, including mannitol and gelatin, are blended, solubilized with water and deaerated before being mixed with the active pharmaceutical ingredient (or "API"), riluzole, which has been milled separately.
Particle size of the API (D50) is less than about 2 microns. The mixture is lyophilized by flash freezing and then freeze-dried. The formulation has good oral palatability.
The effective amount of glutamate modulating agent for the sublingual formulation useful in the present invention to achieve a lower therapeutic dose may be less than that of orally administered agent. Moreover, effective dose of the sublingual formulation of the glutamate modulating agent may be about 1 to 95 % of that of the orally administered agent. To the extent that a sublingual formulation of the immunotherapeutic agent can be made, it may also have improved properties.
The glutamate modulating agent as part of the formulation for treating cancer or symptoms may be dosed at or below about 400 mg/day, at or below about 300 mg/day, at or below about 150 mg/day, at or below about 100 mg/day, at or below about 70 mg/day, at or below about 60 mg/day, at or below about 50 mg/day, at or below about 42.5 mg/day, at or below about 37.5 mg/day at or below about 35 mg/day, at or below about 20 mg/day, at or below about 17.5 mg/day, at or below about 15 mg/day, at or below about 10 mg/day, at or below about 5 mg/day, or at or below about 1 mg/day. In addition, the immunotherapeutic agent should be dosed at about 1-100 mg/kg; for example, 1 mg/kg, 2 mg, kg, 5 mg/kg, 7.5 mg/kg, 10 mg/kg, 20 mg/kg, 25 mg/kg, 50 mg/kg, 75 mg/kg, 100 mg/kg, or any intermediate values. A preferred immunotherapeutic agent is anti-PD-1. Dosing may be daily, alternate days, weekly or having an even higher time separation. In certain circumstances, more often dosing can be used. In addition, the glutamate modulating agent and immunotherapeutic agent can be delivered simultaneously or sequentially.
These and other aspects and features of the invention will be apparent from the drawing and the Detailed Description.
Description of the Drawing
The sole Figure of the Drawing illustrates the results of the test described in Example 1 , testing three levels of a riluzole prodrug (BHV-4157), anti-PD-1 , various combinations of anti-PD-1 and BHV-4157 and a control, to see if the combination modified survival in a glioblastoma animal model.
Detailed Description of the Invention
The following detailed description is provided to aid those skilled in the art in practicing the present invention. Those of ordinary skill in the art may make modifications and variations in the embodiments described herein without departing from the spirit or scope of the present disclosure. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. The terminology used in the description is for describing particular embodiments only and is not intended to be limiting. All publications, patent applications, patents, figures and other references mentioned herein are expressly incorporated by reference in their entirety.
The following terms are used to describe the present invention. In instances where a term is not specifically defined herein, that term is given an art-recognized meaning by those of ordinary skill applying that term in context to its use in describing the present invention.
The articles "a" and "an" as used herein and in the appended claims are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article unless the context clearly indicates otherwise. By way of example, "an element" means one element or more than one element.
The term "riluzole", as used herein, refers to a drug having a chemical structure as follows. It is currently available in the market as RILUTEK®. The term "riluzole" also refers to all prodrugs, enantiomers, or derivatives and its
pharmaceutically acceptable salts.
Figure imgf000008_0001
6-(trifluoromethoxy)benzothiazol-2-amine.
The term "sublingual administration", as used herein, refers to a route of administrating a chemical agent or a drug by placing thereof under a tongue of a subject.
The term "prodrug" as used herein, is a precursor of a drug which may be administered in an altered or less active form. The prodrug may be converted into the active drug form in physiological environments by hydrolysis or other metabolic pathways.
The term "riluzole prodrug" refers to a compound which is a derivative from riluzole with modification therein. A riluzole prodrug may also refer to a compound that is metabolized into an active form of riluzole by the body.
The term "ALS", as used herein, means Amyotrophic Lateral Sclerosis.
The term "immunotherapeutic anti-cancer agent" includes any agent that targets the immune system to result in an anti-cancer therapeutic effects. Such targets and agents include but are not limited to: anti-PD-1, anti-PD-Ll, anti-CTLA4 or other immunotherapy or checkpoint inhibitor targets. Other examples of immuno-oncology targets include: CTLA4, cytotoxic T-lymphocyte-associated antigen 4; Ig, immunoglobulin; LAG3, lymphocyte activation gene 3; mAbs, monoclonal antibodies; PD-1, programmed cell death protein 1; PDL, PD-1 ligand; TIM3, T cell membrane protein 3, CD40L, A2aR, adenosine A2a receptor; B7RP1, B7-related protein 1; BTLA, B and T lymphocyte attenuator; GAL9, galectin 9; HVEM, herpesvirus entry mediator; ICOS, inducible T cell co-stimulator; IL, interleukin; KIR, killer cell immunoglobulin-like receptor; LAG3, lymphocyte activation gene 3; PD-1, programmed cell death protein 1; PDL, PD-1 ligand; TGF , transforming growth factor-β; TIM3, T cell membrane protein 3; and CD27. Other immune targets include: Anti-VEGF-2 monoclonal antibody (Mab), Anti-EGFr Mab, IDOl inhibitor, Anti-B7-H3 Mab, Anti-GITR Mab, Anti-CD 137 Mab, Anti-CD20 Mab, IL- 15 superagonist/IL-15Ra-Fc fusion protein, Anti-CXCR4 Mab, Interleukin-21, Interleukin-21, Anti-KIR Mab, Anti-CD27 Mab, Anti-CSF-IR Mab, Anti-CTLA-4 MAb + GMCSF, Anti-CD30 MAb, Anti-LAG3 Mab, Anti-CD19 Mab, Anti-OX40 Mab, Anti-CD73 Mab, OX40 agonist, or other agents including bi-specific molecules, small molecules targeting the immune system or anti-drug conjugates or vaccines, or nivolumab (Opdivo), pembrolizumab (Keytruda), pidilizumab, ipilimumab (Yervoy), PDR001, MEDI0680, atezolizumab, durvalumab or combinations thereof.
The term "cancer" includes, but is not limited to, the following proliferative diseases: Acute Lymphoblastic Leukemia (ALL), Acute Myeloid Leukemia (AML), Adrenocortical Carcinoms, Childhood cancers, AIDS-Related Cancers, Kaposi Sarcoma, AIDS-Related Lymphoma, Primary CNS Lymphoma, Anal Cancer, Astrocytomas, Atypical Teratoid/Rhabdoid Tumor, Basal Cell Carcinoma, Skin Cancer (Nonmelanoma), Bile Duct Cancer, Bladder Cancer, Bone Cancer, Ewing Sarcoma Family of Tumors, Osteosarcoma and Malignant Fibrous Histiocytoma, Brain Stem Glioma, Atypical Teratoid/Rhabdoid Tumor, Embryonal Tumors, Germ Cell Tumors, Craniopharyngioma, Ependymoma, Breast Cancer, Bronchial Tumors, Burkitt Lymphoma, Non-Hodgkin Lymphoma, Carcinoid Tumor, Gastrointestinal Carcinoma, Cardiac (Heart) Tumors, Primary Lymphoma, Cervical Cancer,
Cholangiocarcinoma, Chordoma, Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), Chronic Myeloproliferative Neoplasms, Colon Cancer, Colorectal Cancer, Craniopharyngioma, Cutaneous T-Cell Lymphoma, Mycosis Fungoides and Sezary Syndrome, Ductal Carcinoma In Situ (DCIS), Embryonal Tumors, Endometrial Cancer, Ependymoma, Esophageal Cancer, Esthesioneuroblastoma, Extracranial Germ Cell Tumor, Extragonadal Germ Cell Tumor, Eye Cancer, Intraocular Melanoma, Retinoblastoma, Fallopian Tube Cancer, Fibrous Histiocytoma of Bone, Malignant, and Osteosarcoma, Gallbladder Cancer, Gastric (Stomach) Cancer, Gastrointestinal Carcinoid Tumor, Gastrointestinal Stromal Tumors (GIST), Germ Cell Tumor, Ovarian, Testicular, Gestational
Trophoblastic Disease, Glioma, Hairy Cell Leukemia, Head and Neck Cancer, Hepatocellular (Liver) Cancer, Histiocytosis, Langerhans Cell, Hodgkin Lymphoma, Hypopharyngeal Cancer, Islet Cell Tumors, Pancreatic Neuroendocrine Tumors, Kaposi Sarcoma, Kidney, Renal Cell, Langerhans Cell Histiocytosis, Laryngeal Cancer, Leukemia, Acute Lymphoblastic (ALL), Acute Myeloid (AML), Chronic Lymphocytic (CLL), Chronic Myelogenous (CML), Hairy Cell, Lip and Oral Cavity Cancer, Liver Cancer (Primary), Lung Cancer, Non-Small Cell, Small Cell, Lymphoma, Hodgkin, Non-Hodgkin, Macroglobulinemia, Waldenstrom, Male Breast Cancer, Melanoma, Merkel Cell Carcinoma, Mesothelioma,Metastatic Squamous Neck Cancer with Occult Primary, Midline Tract Carcinoma Involving NUT Gene, Mouth Cancer, Multiple Endocrine Neoplasia Syndromes, Multiple Myeloma/Plasma Cell Neoplasm, Mycosis Fungoides, Myelodysplasia Syndromes,
Myelodysplastic/Myeloproliferative Neoplasms, Myelogenous Leukemia, Chronic (CML), Myeloid Leukemia, Acute (AML) Myeloma, Multiple, Myeloproliferative Neoplasms, Nasal Cavity and Paranasal Sinus Cancer, Nasopharyngeal Cancer, Neuroblastoma, Non-Hodgkin Lymphoma, Non-Small Cell Lung Cancer, Oral Cancer, Oral Cavity Cancer, Lip and Oropharyngeal Cancer, Osteosarcoma and
Malignant Fibrous Histiocytoma of Bone, Ovarian Cancer, Low Malignant Potential Tumor, Pancreatic Cancer, Pancreatic Neuroendocrine Tumors (Islet Cell Tumors), Papillomatosis, Paraganglioma, Paranasal Sinus and Nasal Cavity Cancer, Parathyroid Cancer, Penile Cancer, Pharyngeal Cancer, Pheochromocytoma, Pituitary Tumor, Plasma Cell Neoplasm/Multiple Myeloma, Pleuropulmonary Blastoma, Pregnancy and Breast Cancer, Primary Central Nervous System (CNS) Lymphoma, Primary Peritoneal Cancer, Prostate Cancer, Rectal Cancer, Renal Cell (Kidney) Cancer, Renal Pelvis and Ureter, Transitional Cell Cancer, Retinoblastoma,
Rhabdomyosarcoma, Salivary Gland Cancer, Rhabdomyosarcoma, Uterine, Small Intestine Cancer, Soft Tissue Sarcoma, Sqamous Cell Carcinoma, Squamous Neck Cancer with Occult Primary, Metastatic, Ttomach (Gastric) Cancer, T-Cell
Lymphoma, Testicular Cancer, Throat Cancer, Thymoma and Thymic Carcinoma, Thyroid Cancer, Transitional Cell Cancer of the Renal Pelvis and Ureter, Unknown Primary, Ureter and Renal Pelvis, Transitional Cell Cancer, Urethral Cancer, Uterine Cancer, Endometrial, Uterine Sarcoma, Vaginal Cancer, Vulvar Cancer, Waldenstrom Macroglobulinemia, and Wilms Tumor.
The term "treatment" as used herein includes any treatment of a condition or disease in a subject, or particularly a human, and may include: (i) preventing the disease or condition from occurring in the subject which may be predisposed to the disease but has not yet been diagnosed as having it; (ii) inhibiting the disease or condition, i.e., arresting its development; relieving the disease or condition, i.e., causing regression of the condition; or (iii) ameliorating or relieving the conditions caused by the disease, i.e., symptoms of the disease. "Treatment," as used herein, could be used in combination with other standard therapies or alone. The term "effective" is used to describe an amount of a compound, composition or component which, when used within the context of its intended use, effects an intended result.
The term "effective amount" refers to that amount which is sufficient to effect treatment, as defined herein, when administered to a subject in need of such treatment. The effective amount will vary depending on the subject and disease state being treated, the severity of the affliction and the manner of administration, and may be determined routinely by one of ordinary skill in the art.
The term "pharmaceutically acceptable salt" is used throughout the specification to describe, where applicable, a salt form of one or more of the compounds or prodrugs described herein which are presented to increase the solubility of the compound in the gastric or gastroenteric juices of the patient's gastrointestinal tract in order to promote dissolution and the bioavailability of the compounds. Pharmaceutically acceptable salts include those derived from
pharmaceutically acceptable inorganic or organic bases and acids, where applicable. Suitable salts include those derived from alkali metals such as potassium and sodium, alkaline earth metals such as calcium, magnesium and ammonium salts, among numerous other acids and bases well known in the pharmaceutical art. Sodium and potassium salts are particularly preferred as neutralization salts of the phosphates according to the present invention. In a preferred embodiment, the description provides pharmaceutically acceptable salts of the modified peptides as described herein, which retain the biological effectiveness and properties of the parent compounds and which are not biologically or otherwise harmful as the dosage administered. The compounds of this invention are capable of forming both acid and base salts by virtue of the presence of amino and carboxy groups respectively.
The term "Cmax", as used herein, refers to a maximum concentration of a drug in blood, serum, a specified compartment or test area of a subject between administration of a first dose and administration of a second dose. The term Cmax could also refer to dose normalized ratios if specified.
The term "Tmax", as used herein, refers to a time or period after
administration of a drug when the maximum concentration (Cmax) is reached in blood, serum, a specified compartment or test area of a subject.
The term "AUC" (area under the curve), as used herein, refers to a total amount of drug absorbed or exposed to a subject. Generally, AUC may be obtained from mathematical method in a plot of drug concentration in the subject over time until the concentration is negligible. The term "AUC" (area under the curve) could also refer to partial AUC at specified time intervals (as may be the case with sublingual absorption which would increase AUC at earlier time intervals).
The invention relates to a combination therapy utilizing an immunotherapeutic agent and a glutamate modulating agent. The combination of these two drugs may be administered in a single dose as combined product, administered simultaneously using the same or distinct formats, or administered sequentially using the same or different forms of delivery. For example, if the immunotherapeutic agent and the glutamate modulating agent can both be made into a tablet or part of a sublingual form, they can be administered together. Similarly, if the immunotherapeutic agent can only be administered by injection (bolus or intravenous), and the glutamate modulating agent can be administered in the same format, this could also be used for simultaneous or sequential administration. However, if the immunotherapeutic agent can only be delivered by injection (for example, if it is an antibody), and the glutamate modulating agent can be delivered as a tablet or sublingually, delivery of the two agents can take place by differing formats.
Some of the glutamate modulating agents can be administered sublingually. PCT Application No. PCT/US2015/061106 and PCT Application No.
PCT/US2015/061114 describe a sublingual formulation of riluzole, a preferred glutamate modulating agent. The sublingual formulation may be administered in an effective amount to a subject in need thereof. The subject may be an animal or human.
For use with the current invention, the glutamate modulating agent or its pharmaceutically acceptable salts thereof may be formulated in a pharmaceutical composition suitable for sublingual administration. In some circumstances, the immunotherapeutic agent may also be formulated as a sublingual, although injection is more standard.
Glutamate modulating agents such as riluzole and the pharmaceutically acceptable salts thereof can be formulated using pharmaceutically acceptable carriers well known in the art into dosages suitable for sublingual or buccal administration. Such carriers enable the glutamate modulating agent for sublingual administration to be formulated in dosage forms such as tablets, powders, pills, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for sublingual absorption by a subject to be treated. These carriers may be, but not limited to, selected from sugars, starches, cellulose and its derivatives, malt, gelatin, talc, calcium sulphate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffered solutions, emulsifiers, isotonic saline, pyrogen-free water and combinations thereof. In particular, any form of substance may be accepted to sublingual administration if it dissolves easily in saliva.
The sublingually administered chemical agent or the drug can diffuse into capillaries through mucous membrane under the tongue, and then enter venous circulation of the subject. As such, sublingual administration may have advantages over oral administration as allowing for direct or faster entry to venous circulation, without risks of degradation in gastrointestinal tract, alteration by drug metabolism in liver and the like. Various drugs in the market are designed for sublingual administration. Riluzole is generally used to treat amyotrophic lateral sclerosis (ALS). However, other uses have been found, and in particular, riluzole or prodrugs of riluzole or pharmaceutically acceptable salts thereof is subjected to a sublingual administration for the treatment of other disorders, including cancer.
The pharmaceutical composition may include an approved pharmaceutical ingredient, i.e., riluzole, in an effective amount to achieve their intended purpose. For example, the dose of the glutamate modulating agent administered sublingually to the subject should be sufficient to provide a beneficial response in the subject over time such as reduction in symptoms in conjunction with the immunotherapeutic agent. The combination may have synergistic effects.
The quantity of the glutamate modulating agent and the quantity of the immunotherapeutic agent to be administered may depend on the subject to be treated inclusive of the age, sex, weight and general health condition thereof. In this regard, precise amounts of the agent(s) for administration will depend on the judgment of the practitioner. In determining the effective amount of the glutamate modulating agent and immunotherapeutic agent to be administered in the treatment or reducing of the conditions associated with the symptoms and disorders, the physician may evaluate clinical factors including symptoms severity or progression of the disorder. In some conditions, a rapid absorption of the glutamate modulating agent or
immunotherapeutic agent may be desirable. In any event, those of skill in the art may readily determine suitable dosages of the chemical agents of the invention.
The pharmaceutical composition also includes other pharmaceutically acceptable carriers and/or excipients such as binders, lubricants, diluents, coatings, disintegrants, barrier layer components, glidants, coloring agents, solubility enhancers, gelling agents, fillers, proteins, co-factors, emulsifiers, solubilizing agents, suspending agents and mixtures thereof. A skilled artisan in the art would know what other pharmaceutically acceptable carriers and/or excipients could be included in the formulations according to the invention. The choice of excipients would depend on the characteristics of the compositions and on the nature of other pharmacologically active compounds in the formulation. Appropriate excipients are known to those skilled in the art (see Handbook of Pharmaceutical Excipients, fifth edition, 2005 edited by Rowe et al, McGraw Hill) and have been utilized to yield a novel sublingual formulation with unexpected properties.
In addition, the pharmaceutical composition for sublingual use can be obtained by combining an approved pharmaceutical ingredient, i.e., riluzole, with further excipients, with optionally processing to obtain dosage forms such as tablets, powders, pills, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for sublingual absorption by a subject to be treated. Suitable excipients may be, but not limited to, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropyl methyl-cellulose, sodium carboxymethylcellulose, and/or polyvinyl-pyrrolidone (PVP). If desired,
disintegrating agents may be combined as well, and exemplary disintegrating agents may be, but not limited to, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. The compositions may be prepared by any of the methods of pharmacy but all methods include the step of bringing into association one or more chemical agents as described above with the carrier which constitutes one or more necessary ingredients. In general, the pharmaceutical compositions of the present invention may be manufactured in conventional methods known in the art, for example, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, lyophilizing processes and the like.
The sublingual formulation useful in the combination product of the invention may be prepared in a form of an orally dissolving or disintegrating tablet (ODT). The ODT as used herein may be prepared by mixing the glutamate modulating agent and/or the immunotherapeutic agent with water-soluble diluents and compressed in a tablet. A suspension comprising the active product may be prepared with appropriate excipients and the suspension may be dispensed into blister packs and freeze-dried. An exemplary freeze-dried preparation platform that could be used for the ODT is the ZYDIS® (Catalent, Somerset, NJ, USA) formulation. In particular, the excipients, including water, are blended and the glutamate modulating agent is separately milled to size and mixed with the excipients. The suspension then undergoes lyophilisation by flash freezing and freeze drying. Other methods of preparing ODTs may be used without limitation, and detailed description of general methods thereof have been disclosed, for example, in U.S. Pat. No 5,631,023; 5,837,287; 6 ,149,938; 6,212,791; 6,284,270; 6,316,029; 6,465,010; 6,471,992; 6,471,992; 6,509,040; 6,814,978;
6,908,626; 6,908,626; 6,982,251; 7,282,217; 7,425,341 ; 7,939,105; 7,993,674;
8,048,449; 8,127,516; 8,158,152; 8,221,480; 8,256,233; and 8,313,768, each of which is incorporated herein by reference in its entirety.
The sublingual formulation useful in the invention may comprise the glutamate modulating agent or an effective amount of a glutamate modulating agent prodrug. Similarly, in certain circumstances, the immunotherapeutic agent may be made into a prodrug. The prodrug may be similar or less active form of the active. The prodrug may have improved physiochemical, physiological pharmacokinetic or therapeutical characteristics when administered sublingually. The prodrug may reduce side effects when orally or sublingually administered.
The clinical or therapeutic effect of the compound, or a subportion of the final product, sublingually formulated may have an improved pharmacokinetic profile for the pharmaceutical agent as measured by standard testing parameters. When the glutamate modulating agent or the total composition is administered sublingually, the Tmax, Cmax and AUC of the drug may be improved compared to the same dose of the orally administered version of the same compound. For example, the sublingual formulation of the glutamate modulating agent may have a greater Cmax than the orally administered glutamate modulating agent to provide a therapeutically beneficial effect. The sublingual formulation of the glutamate modulating agent may have an earlier or lesser Tmax than the orally administered glutamate modulating agent to provide a therapeutically beneficial effect and in some instances, a more rapid therapeutic effect. Alternatively, the sublingual formulation of the glutamate modulating agent may have a greater AUC per milligram of the agent than the orally administered glutamate modulating agent. In addition, as the glutamate modulating agent may make the immunotherapeutic agent more effective, lesser amounts of the immunotherapeutic agent may be needed to achieve the same results, with a lessening of the inherent side effects.
The invention provides a method of treating a disease such as cancer. The method comprises administering sublingually an effective amount of glutamate modulating agent or pharmaceutically acceptable salts thereof and an anti-cancer agent, preferably an immunotherapeutic agent, or pharmaceutically acceptable salts thereof to a subject in need thereof.
Identifying the subject in need of such treatment can be in the judgment of the subject or a health care professional and can be subjective (e.g., opinion) or objective (e.g., measurable by a test or diagnostic method). The identified subject may be an animal or human in need thereof, particularly a human. Such treatment will be suitably administered to subjects, particularly humans, suffering from the disease.
The effective amount of the treatment will vary depending on the subject and disease state being treated, the severity of the affliction and the manner of
administration, and may be determined routinely by one of ordinary skill in the art.
The therapeutic effect of the combination product, particularly as it applies to treating symptoms, may be evident to occur within about a few minutes to about an hour after administration thereof. In particular, the therapeutic effect may begin within about 1 minute, within about 2 minutes, within about 3 minutes, within about 4 minutes, within about 5 minutes, within about 6 minutes, within about 7 minutes, within about 8 minutes, within about 9 minutes, within about 10 minutes, within about 11 minutes, within about 12 minutes, within about 13 minutes, within about 14 minutes, within about 15 minutes, within about 16 minutes, within about 17 minutes, within about 18 minutes, within about 20 minutes, within about 60 minutes, or within about 90 minutes after administration. However, long term cure or amelioration of the disease may not occur for weeks or months after administration.
The effects on the symptoms may be maintained for about 1 hour, for about 2 hours, for about 3 hours, for about 4 hours, for about 5 hours, for about 6 hours m for about 7 hours, for about 8 hours, for about 9 hours, for about 10 hours, for about 12 hours, for about 14 hours, for about 16 hours, for about 18 hours, for about 20 hours, for about 22 hours, for about 24 hours, for about 2 days, or for about 3 days or more after administration thereof. Hopefully, once the long term effects on the disease state is achieved, the disease, and the symptoms, will be eliminated permanently. The effective amount or dose of glutamate modulating agent for sublingual administration may be less than that of orally administered agent. In particular, the effective dose in sublingual administration of the glutamate modulating agent, such as riluzole, may be of about 1-95 % of the dose of the orally administered agent itself. In addition, similar reduction in the amount of the immunotherapeutic agent may be achieved by administration of the glutamate modulating agent itself, by any mode of administration.
Optional dosage frequencies include once a day, twice a day, three times a day, four times a day, once every other day, once a week, twice a week, three times a week, four times a week, once every two weeks, once or twice monthly, and the like.
Glutamate modulating agents can be used as is or may be in the form of prodrugs. Prodrugs of riluzole are described in United States Patent Application Serial No. 14/385,551, United States Patent Application Serial No. 14/410,647, PCT Application Serial No. PCT/US2016/019773 and PCT Application Serial
No.PCT/US2016/019787. The preferred riluzole prodrugs have the structure:
Figure imgf000017_0001
including enantiomers, diasteroemers, hydrates, solvates, pharmaceutically acceptable salts, and complexes thereof, wherein:
P 23 is selected from the group consisting H, CH3, CH2CH3, CH2CH2CH3, CH2CCH, CH(CH3)2, CH2CH(CH3)2, CH(CH3)CH2CH3, CH2OH, CH2OCH2Ph,
CH2CH2OCH2Ph, CH(OH)CH3, CH2Ph, CH2(cyclohexyl), CH2(4-OH-Ph),
(CH2)4NH2, (CH2)3NHC(NH2)NH, CH2(3 -indole), CH2(5-imidazole), CH2CO2H, CH2CH2CO2H, CH2CONH2, and CH2CH2CONH2.
A preferred prodrug of riluzole has the following formula:
Figure imgf000017_0002
Those skilled in the art will recognize that similar or variant prodrugs can be made from other glutamate modulating agents. Such agents are useful as part of the combination of the present invention.
EXAMPLE
The following example illustrates the invention and is not intended to limit the scope of the invention.
In this Example, the effects of the combination of a glutamate modulator, BHV-4157, in combination with an immunotherapeutic agent, anti-PD-1, were compared to either alone in a glioma model substantially as decribed in Zeng, J., et al., Int J Radiat Oncol Biol Phys., 2013 June 1; 86(2):343-349, portions of which are reproduced below. Cells
GL261-Luc cells are grown in Dulbecco's Modified Eagle Medium (DMEM) + 10% fetal bovineserum + 1% penicillinstreptomycin at 37°C in a humidified incubator maintained at 5% CO and 5% O2 (Gibco). Tumor Model
Female C57BL/6J mice (Harlan), 4 to 6 weeks old or 6 to 8 weeks old, are used for orthotopic glioma experiments as described in Sonabend AM, Velicu S, Ulasov IV, et al. A safety and efficacy study of local delivery of interleukinl2 transgene by PPC polymer in a model of experimental glioma. Anticancer Drugs. 2008;19: 133-142. To establish syngeneic gliomas, 130,000 GL261-Luc cells are stereotactically injected in a Ιμί volume into the left striatum over 1 minute into the following coordinates: 1 mm anterior, 1 mm lateral from bregma, and 3 mm deep from the cortical surface. Tumor burden is monitored by luciferase imaging on days 7, 21 and 35 after implantation, and the mice are randomly allocated into treatment arms based on tumor radiance, so that the average tumor radiance in each group is roughly equivalent. The animals are euthanized when they show predetermined signs of neurologic deficits (failure to ambulate, weight loss >20%body mass, lethargy, hunched posture). The tumor take rate is 100%. Each arm has 6 to 10 mice in survival experiments. All experiments are repeated at least in triplicate. Anti-PD-1 antibodies
Hamster antimurine PD-1 monoclonal antibody producing hybridoma G4 are used to produce antibodies as described in Hirano F, Kaneko K, Tamura H, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005;65: 1089-1096.
Specific Protocol
Female C57BL/6J mice, 4 to 6 weeks old, were implanted intrancranially in the left striatum with 130,000 GL261 cells each. The mice were housed and maintained according to the institutional Animal Care and Use Committee protocol in the Johns Hopkins University Animal Facility. The mice were imaged by
bioluminescent iVIS© imaging (Perkin Elmer) at day 7, 21 , and 35 to assess tumor burden and randomly assigned to groups, 10 mice per arm, as follows:
Control
anti-PD-l
Trigriluzole 15 mg/kg
Trigriluzole 30 mg kg
Trigriluzole 45 mg/kg
anti-PD-l + Trigriluzole 15 mg/kg
anti-PD-l + Trigriluzole 30 mg/kg
anti-PD-l + Trigriluzole 45 mg/kg
The protocol is shown in the following illustration:
Day 0 represents the date of intracranial implantation. Control arm 1 received no treatment. Control arm 2 received aPD-1 alone at a dose of 200 ug/animal via intraperitoneal injection on days 10, 12, 14. Control arms 3, 4 and 5 received BHV- 4157 alone at doses of 15, 30 and 45 mg/kg (respectively) via intraperitoneal injection daily beginning on day 10. Control arms 6, 7 and 8 received BHV-4157 at doses of 15, 30 and 45 mg/kg (respectively) via intraperitoneal injection daily beginning on day 10 and aPD-1 at a dose of 200 ug/animal via intraperitoneal injection on days 10, 12, 14.
The treatment was terminated when mice showed no tumor burden via WIS imaging. Animals were euthanized according to humane endpoints including central nervous system disturbances, hunched posture, lethargy, weight loss, and inability to ambulate.
The purpose of the experiment was to see if the combination therapy was provided a benefit over either therapy alone. The results are shown in Figure 1. As is evident from Figure 1, the combination therapy is substantially better than any of the individual therapies and that the effects are not merely additive but appear synergistic. Thus, it appears that the glutamate modulators effect on glutamate/glutamine metabolism weakens the tumor cells and makes the anti-PD-1 antibody more effective. Quite surprisingly, in accordance with the present invention, the percentage survival of mice at about 30, 40 and 60 days after implantation was about 2 times, or greater, the percent survival for the mice treated with the glutamate modulator in combination with the immunotherapeutic anti-cancer agent as compared to the immunotherapeutic anti-cancer agent alone. Table 1 below shows data from Example 1.
TABLE 1
Figure imgf000021_0001
The term "Mouse Survival Ratio", also referred to as "MSRx" refers to a value calculated by dividing: (i) the percentage survival of mice treated with an immunotherapeutic anti-cancer agent plus a glutamate modulating agent, by (ii) the percentage survival of mice treated with an immunotherapeutic anti-cancer agent alone, in accordance with the procedure set forth in Example 1 hereof at a time period of "x" number of days after implantation of the tumor into the mice. Thus, MSR60 refers to the Mouse Survival Ratio at a time of 60 days after tumor implantation.
From Table 1, it can be seen that by day 26, the mice in Arm 1 (Control) had 0% survival, the mice in Arm 2 (PD-1) had 50% survival and the mice in Arms 6, 7 and 8 had at least 70 to 80% survival. Accordingly, at day 26, the Mouse Survival Ratio (MSR26) was about 1.4 to 1.6 (i.e., 70/50 and 80/50). At day 28, the mice in Arm 1 (Control) had 0% survival, the mice in Arm 2 (PD-1) had 30% survival and the mice in Arms 6, 7 and 8 had at least 60 to 80% survival. Accordingly, at day 28, the Mouse Survival Ratio (MSR28) was about 2.0 to 2.6 (i.e., 60/30 and 80/30). At day 60, the mice in Arm 1 (Control) had 0% survival, the mice in Arm 2 (PD-1) had 30% survival and the mice in Arms 6, 7 and 8 had 60 to 70% survival. Accordingly, at day 60, the Mouse Survival Ratio (MSR60) was about 2.0 to 2.3 (i.e., 60/30 and 70/30). Preferably, in accordance with the present invention, the Mouse Survival Ratio is at least 1.4, more preferably at least 1.6 when measured at 26 days after tumor implantation (MSR26). Preferably, in accordance with the present invention, the Mouse Survival Ratio is at least 2.0, more preferably at least 2.6 when measured at 28 days after tumor implantation (MSR28). Preferably, in accordance with the present invention, the Mouse Survival Ratio is at least 2.0, more preferably at least 2.3 when measured at 60 days after tumor implantation (MSR60). Preferably, in accordance with the present invention, the Mouse Survival Ratio measured at a time when the untreated mice reach 0% survival, or thereafter until a time of 60 days after tumor implantation, is at least 1.4, at least 1.6, at least 2.0, at least 2.3 or at least 2.6.
Typically, combination therapy, i.e., an immunotherapeutic anti-cancer agent and a glutamate modulating agent, in accordance with the present invention will provide a Mouse Survival Ratio of at least 2.0, more typically at least 2.3 (measured at day 60, MSR60).
The results are shown in Figure 1. As is evident from Figure 1, the combination therapy is substantially better than any of the individual therapies and that the effects are not merely additive but appear synergistic. Thus, it appears that the glutamate modulators effect on glutamate/glutamine metabolism weakens the tumor cells and makes the anti-PD-1 more effective. A variety of solid malignancies have been shown to overexpress phosphate-dependent glutaminase (GLS), which converts glutamine to glutamate further emphasizing the role of glutamine in cancer metabolism. However, glutamate is a key nitrogen "waste" bank and critical in a variety of cellular metabolic pathways. As such, reduction in glutamine/glutamate levels to immune cells may reduce proliferative and effector function, limiting an anti-tumor immune mediated response. While this effect is clear for GLS producing tumor cells, glutamate receptors are found on a number of other tumor cells and it is believed that this combination therapy could be effective for those cells as well.
The entire contents of all patents, published patent applications and other references cited herein are hereby expressly incorporated herein in their entireties by reference.
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific procedures described herein. Such equivalents are considered to be within the scope of this invention and are covered by the following claims.

Claims

CLAIMS What is claimed is:
1. A method of treating cancer by administering a combination therapy to a patient in need thereof comprising of an effective amount of riluzole and a checkpoint inhibitor.
2. The method of claim 1 wherein riluzole and the checkpoint inhibitor are administered either concurrently or sequentially in time.
3. The method of claim 1 wherein said checkpoint inhibitor targets PD-1 , PD-L1 or CTLA-4.
4. The method of claim 1 wherein said checkpoint inhibitor is selected from the group consisting of nivolumab, pembrolizumab, pidilizumab, ipilimumab, PDROOl, MEDI0680, atezolizumab, durvalumab and combinations thereof.
5. The method of claim 1 wherein said riluzole is in the form of an analog or prodrug of riluzole, or pharmaceutically acceptable salts, solvates, anomers, enantiomers, or hydrates thereof.
6. The method of claim 1 wherein said combination of riluzole and checkpoint inhibitor is administered in combination or sequentially with other anti-cancer standard of care treatments.
7. The method of claim 1 wherein said combination is riluzole and an anti-PDl or anti- PD-L1 and the cancer treated is glioblastoma.
8. The method of claim 1 wherein said combination is riluzole and an anti-PDl or anti- PD-L1 and the cancer treated is melanoma.
9. The method of claim 5 wherein the prodrug has the following formula:
Figure imgf000025_0001
10. The method of claim 1 wherein the riluzole and the checkpoint inhibitor are capable of providing a Mouse Survival Ratio of at least 2,0 at day 60 (MSR60).
11. The method of claim 4 wherein the riluzole and the checkpoint inhibitor are capable of providing a Mouse Survival Ratio of at least 2.0 at day 60 (MS 50).
12. The method of claim 5 wherein the prodrug of riluzole and the checkpoint inhibitor are capable of providing a Mouse Survival Ratio of at least 2.0 at day 60 (MSR60).
13. The method of claim 9 wherein the prodrug of riluzole and the checkpoint inhibitor are capable of providing a Mouse Survival Ratio of at least 2.0 at day 60 (MSR60).
PCT/US2017/033688 2016-05-20 2017-05-19 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers WO2017201501A1 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
RS20220165A RS62935B1 (en) 2016-05-20 2017-05-19 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers
EP21207246.6A EP4019019B1 (en) 2016-05-20 2017-05-19 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers
US16/302,284 US11400155B2 (en) 2016-05-20 2017-05-19 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers
SI201731061T SI3458053T1 (en) 2016-05-20 2017-05-19 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers
JP2018560520A JP7169195B2 (en) 2016-05-20 2017-05-19 Combination of riluzole, riluzole prodrugs or riluzole analogues with immunotherapy to treat cancer
DK17800310.9T DK3458053T3 (en) 2016-05-20 2017-05-19 USE OF RILUZOLE, RILUZOLE PRODUCT OR RILUZOLE ANALOGIES WITH CANCER TREATMENT FOR CANCER TREATMENT
PL17800310T PL3458053T3 (en) 2016-05-20 2017-05-19 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers
LTEPPCT/US2017/033688T LT3458053T (en) 2016-05-20 2017-05-19 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers
EP17800310.9A EP3458053B1 (en) 2016-05-20 2017-05-19 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers
ES17800310T ES2905823T3 (en) 2016-05-20 2017-05-19 Use of riluzole, riluzole prodrugs, or riluzole analogs with immunotherapies to treat cancers
HRP20220237TT HRP20220237T1 (en) 2016-05-20 2017-05-19 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers
CY20221100131T CY1124999T1 (en) 2016-05-20 2022-02-16 USE OF RILUSOLE, RILUSOLE PRODRUGS, OR RILUSOLE ANALOGUES WITH IMMUNE THERAPIES FOR THE TREATMENT OF CANCER
US17/878,121 US20230310595A1 (en) 2016-05-20 2022-08-01 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers
JP2022173334A JP2023012508A (en) 2016-05-20 2022-10-28 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662339433P 2016-05-20 2016-05-20
US62/339,433 2016-05-20

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/302,284 A-371-Of-International US11400155B2 (en) 2016-05-20 2017-05-19 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers
US17/878,121 Continuation US20230310595A1 (en) 2016-05-20 2022-08-01 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers

Publications (1)

Publication Number Publication Date
WO2017201501A1 true WO2017201501A1 (en) 2017-11-23

Family

ID=60326148

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2017/033688 WO2017201501A1 (en) 2016-05-20 2017-05-19 Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers
PCT/US2017/033690 WO2017201502A1 (en) 2016-05-20 2017-05-19 Use of glutamate modulating agents with immunotherapies to treat cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2017/033690 WO2017201502A1 (en) 2016-05-20 2017-05-19 Use of glutamate modulating agents with immunotherapies to treat cancer

Country Status (25)

Country Link
US (3) US20190175731A1 (en)
EP (3) EP3458054B1 (en)
JP (4) JP7224186B2 (en)
KR (2) KR102369735B1 (en)
CN (2) CN115350263A (en)
AU (2) AU2017266951B2 (en)
BR (1) BR112018073781A2 (en)
CA (1) CA3025019A1 (en)
CY (1) CY1124999T1 (en)
DK (1) DK3458053T3 (en)
EA (1) EA201892587A1 (en)
ES (2) ES2905823T3 (en)
HR (1) HRP20220237T1 (en)
HU (1) HUE058184T2 (en)
IL (2) IL292302B2 (en)
LT (1) LT3458053T (en)
MX (2) MX2018013868A (en)
PH (1) PH12018502409A1 (en)
PL (1) PL3458053T3 (en)
PT (1) PT3458053T (en)
RS (1) RS62935B1 (en)
SG (1) SG11201809669RA (en)
SI (1) SI3458053T1 (en)
WO (2) WO2017201501A1 (en)
ZA (1) ZA201807398B (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019164861A1 (en) * 2018-02-21 2019-08-29 AI Therapeutics, Inc. Combination therapy with apilimod and glutamatergic agents
US20210236471A1 (en) * 2015-03-03 2021-08-05 Biohaven Therapeutics Ltd. Riluzole prodrugs and their use
US11168144B2 (en) 2017-06-01 2021-11-09 Cytomx Therapeutics, Inc. Activatable anti-PDL1 antibodies, and methods of use thereof
CN114222729A (en) * 2019-06-12 2022-03-22 范德比尔特大学 Amino acid transport inhibitors and uses thereof
WO2023202652A1 (en) * 2022-04-21 2023-10-26 Jacobio Pharmaceuticals Co., Ltd. Pharmaceutical combination and use thereof
US11957688B2 (en) 2022-07-12 2024-04-16 OrphAl Therapeutics Inc. Combination therapy with apilimod and glutamatergic agents

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013138753A1 (en) * 2012-03-16 2013-09-19 Fox Chase Chemical Diversity Center, Inc. Prodrugs of riluzole and their method of use
CN107849144B (en) 2015-05-29 2021-09-17 艾吉纳斯公司 anti-CTLA-4 antibodies and methods of use thereof
JP7014706B2 (en) 2015-07-13 2022-02-01 サイトメックス セラピューティクス インコーポレイテッド Anti-PD-1 antibody, activating anti-PD-1 antibody, and how to use it
WO2017062354A1 (en) 2015-10-05 2017-04-13 Calithera Biosciences, Inc. Combination therapy with glutaminase inhibitors and immuno-oncology agents
MX2018013868A (en) * 2016-05-20 2019-02-21 Biohaven Pharm Holding Co Ltd Use of glutamate modulating agents with immunotherapies to treat cancer.
MA46529A (en) 2016-10-11 2019-08-21 Agenus Inc ANTI-LAG-3 ANTIBODIES AND PROCESSES FOR USE
IL266918B2 (en) 2016-12-07 2024-03-01 Agenus Inc Anti-ctla-4 antibodies and methods of use thereof
JP7437301B2 (en) 2017-08-25 2024-02-22 ファイヴ プライム セラピューティクス インク B7-H4 antibody and its usage
WO2019089952A1 (en) * 2017-11-03 2019-05-09 Calithera Biosciences, Inc. Conjoint therapy with glutaminase inhibitors
EA202091810A1 (en) 2018-03-02 2021-01-29 Файв Прайм Терапьютикс, Инк. ANTIBODIES TO B7-H4 AND METHODS OF THEIR APPLICATION
US20210290599A1 (en) * 2018-07-22 2021-09-23 Biohaven Therapeutics Ltd. Use of riluzole prodrugs to treat alzheimer's disease
US20220081679A1 (en) * 2019-01-24 2022-03-17 University Of Cincinnati Autologous tumor organoid and immune cell co-cultures and methods of use as predictive models for pancreatic cancer treatment
CN110646557A (en) * 2019-10-12 2020-01-03 北京航空航天大学 Urine metabolic marker of glioblastoma patient carrying IDH gene mutation and application thereof
JP7431412B2 (en) 2019-12-23 2024-02-15 三菱マテリアル株式会社 How to separate covered wires
IL296508A (en) * 2020-03-17 2022-11-01 Ohio State Innovation Foundation Designer extracellular vesicles for treating excitotoxicity
KR102441650B1 (en) * 2020-07-31 2022-09-08 서울대학교병원 Method for providing effective dose information on anticancer medicine in multidrug chemotherapy
WO2022197767A1 (en) * 2021-03-17 2022-09-22 Memorial Sloan-Kettering Cancer Center Methods for improving survival in lung cancer patients via ketamine oncoprotection
WO2023003972A1 (en) * 2021-07-21 2023-01-26 Viracta Subsidiary, Inc. Cancer treatment combinations
CN116492322A (en) * 2023-05-10 2023-07-28 徐州医科大学 Application of small molecule blocker MEM in preparation of tumor treatment drugs

Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5631023A (en) 1993-07-09 1997-05-20 R.P. Scherer Corporation Method for making freeze dried drug dosage forms
US5837287A (en) 1994-10-28 1998-11-17 R P Scherer Corporation Process for preparing solid pharmaceutical dosage forms
US6149938A (en) 1997-07-25 2000-11-21 Elan Pharma International Limited Process for the preparation of a granulate suitable to the preparation of rapidly disintegrable mouth-soluble tablets and compositions obtained thereby
US6212791B1 (en) 1993-10-01 2001-04-10 R.P. Scherer Corporation Method of applying indicia to a fast-dissolving dosage form
US6284270B1 (en) 1999-08-04 2001-09-04 Drugtech Corporation Means for creating a mass having structural integrity
US6316029B1 (en) 2000-05-18 2001-11-13 Flak Pharma International, Ltd. Rapidly disintegrating solid oral dosage form
US6471992B1 (en) 1997-02-20 2002-10-29 Therics, Inc. Dosage form exhibiting rapid disperse properties, methods of use and process for the manufacture of same
US6509040B1 (en) 2001-06-22 2003-01-21 R.P. Scherer Corporation Fast dispersing dosage forms essentially free of mammalian gelatin
US6814978B2 (en) 2000-12-29 2004-11-09 Mcneil-Ppc, Inc. Process for preparing a soft tablet
US6908626B2 (en) 2001-10-12 2005-06-21 Elan Pharma International Ltd. Compositions having a combination of immediate release and controlled release characteristics
US6982251B2 (en) 2000-12-20 2006-01-03 Schering Corporation Substituted 2-azetidinones useful as hypocholesterolemic agents
US7282217B1 (en) 2003-08-29 2007-10-16 Kv Pharmaceutical Company Rapidly disintegrable tablets
US7939105B2 (en) 1998-11-20 2011-05-10 Jagotec Ag Process for preparing a rapidly dispersing solid drug dosage form
US7993674B2 (en) 2002-02-13 2011-08-09 Weibel Michael K Drug dose-form and method of manufacture
US8048449B2 (en) 2005-12-27 2011-11-01 Jubilant Organosys Ltd. Mouth dissolving pharmaceutical composition and process for preparing the same
US8127516B2 (en) 2007-06-27 2012-03-06 Hanmi Pharm. Co., Ltd. Method for preparing rapidly disintegrating formulation for oral administration and apparatus for preparing and packing the same
US8158152B2 (en) 2005-11-18 2012-04-17 Scidose Llc Lyophilization process and products obtained thereby
US8221480B2 (en) 2008-10-31 2012-07-17 The Invention Science Fund I, Llc Compositions and methods for biological remodeling with frozen particle compositions
US8256233B2 (en) 2008-10-31 2012-09-04 The Invention Science Fund I, Llc Systems, devices, and methods for making or administering frozen particles
US8313768B2 (en) 2009-09-24 2012-11-20 Mcneil-Ppc, Inc. Manufacture of tablet having immediate release region and sustained release region
WO2013138753A1 (en) 2012-03-16 2013-09-19 Fox Chase Chemical Diversity Center, Inc. Prodrugs of riluzole and their method of use
US20150148329A1 (en) * 2012-06-23 2015-05-28 Fox Chase Chemical Diversity Center, Inc. Pro-drugs of riluzole and their method of use for the treatment of amyotrophic lateral sclerosis
US20160019787A1 (en) 2011-10-20 2016-01-21 Apple Inc. Method for Locating a Vehicle
WO2016073759A1 (en) * 2014-11-05 2016-05-12 The Regents Of The University Of California Combination immunotherapy

Family Cites Families (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7812A (en) 1850-12-03 Nail-plate feeder and turner
CA209066A (en) 1921-03-01 Tschirner Frederick Conversion of potassium to soluble state
US135A (en) 1837-03-03 Jesse j
US6355476B1 (en) 1988-11-07 2002-03-12 Advanced Research And Technologyinc Nucleic acid encoding MIP-1α Lymphokine
US6303121B1 (en) 1992-07-30 2001-10-16 Advanced Research And Technology Method of using human receptor protein 4-1BB
US6362325B1 (en) 1988-11-07 2002-03-26 Advanced Research And Technology Institute, Inc. Murine 4-1BB gene
DK0590058T3 (en) 1991-06-14 2004-03-29 Genentech Inc Humanized heregulin antibody
US5851795A (en) 1991-06-27 1998-12-22 Bristol-Myers Squibb Company Soluble CTLA4 molecules and uses thereof
US6051227A (en) 1995-07-25 2000-04-18 The Regents Of The University Of California, Office Of Technology Transfer Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
US7423125B2 (en) 1995-08-01 2008-09-09 Vegenics Limited Antibodies to VEGF-C
US6100071A (en) 1996-05-07 2000-08-08 Genentech, Inc. Receptors as novel inhibitors of vascular endothelial growth factor activity and processes for their production
WO1998016249A1 (en) 1996-10-11 1998-04-23 Bristol-Myers Squibb Company Methods and compositions for immunomodulation
US20020032315A1 (en) 1997-08-06 2002-03-14 Manuel Baca Anti-vegf antibodies
ES2273415T3 (en) 1997-04-07 2007-05-01 Genentech, Inc. ANTI-VEGF ANTIBODIES.
EE05627B1 (en) 1998-12-23 2013-02-15 Pfizer Inc. Human monoclonal antibodies to CTLA-4
BRPI0017590B8 (en) 1999-06-25 2021-05-25 Genentech Inc maytansinoid conjugate - anti-erbb antibody, and pharmaceutical formulation
KR20020047132A (en) 1999-08-24 2002-06-21 메다렉스, 인코포레이티드 Human ctla-4 antibodies and their uses
US7605238B2 (en) 1999-08-24 2009-10-20 Medarex, Inc. Human CTLA-4 antibodies and their uses
EP1261376A1 (en) 2000-01-27 2002-12-04 Genetics Institute, LLC Antibodies against ctla4(cd152), conjugates comprising same, and uses thereof
AU2003213687A1 (en) 2002-03-04 2003-09-22 Imclone Systems Incorporated Human antibodies specific to kdr and uses thereof
JP2006500921A (en) 2002-07-30 2006-01-12 ブリストル−マイヤーズ スクイブ カンパニー Humanized antibody against human 4-1BB
EP1639013B1 (en) 2003-07-02 2012-09-12 Innate Pharma Pan-kir2dl NK-receptor antibodies and their use in diagnostic and therapy
EP2292264A3 (en) 2003-07-24 2012-12-19 Innate Pharma Methods and compositions for increasing the efficiency of therapeutic antibodies using NK cell potentiating compounds
US7288638B2 (en) 2003-10-10 2007-10-30 Bristol-Myers Squibb Company Fully human antibodies against human 4-1BB
WO2006003179A2 (en) 2004-07-01 2006-01-12 Novo Nordisk A/S Antibodies binding to receptors kir2dl1, -2, 3 but not kir2ds4 and their therapeutic use
CA2578066C (en) 2004-08-26 2011-10-11 Pfizer Inc. Enantiomerically pure aminoheteroaryl compounds as protein kinase inhibitors
CN101018780B (en) 2004-08-26 2012-01-11 辉瑞大药厂 Pyrazole-substituted aminoheteroaryl compounds as protein kinase inhibitors
DK1819358T3 (en) 2004-11-18 2014-10-27 Imclone Llc Antibodies to Vascular Endothelial Growth Factor Receptor-1
PL1836225T3 (en) 2005-01-06 2012-05-31 Novo Nordisk As Kir-binding agents and methods of use thereof
EP3072522B1 (en) 2005-01-06 2019-04-24 Novo Nordisk A/S Anti-kir combination treatments and methods
DK2343320T3 (en) 2005-03-25 2018-01-29 Gitr Inc ANTI-GITR ANTIBODIES AND APPLICATIONS THEREOF
DK2161336T4 (en) 2005-05-09 2017-04-24 Ono Pharmaceutical Co Human monoclonal antibodies for programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapies
EP1907424B1 (en) 2005-07-01 2015-07-29 E. R. Squibb & Sons, L.L.C. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
EP1744020A1 (en) 2005-07-14 2007-01-17 Siemens Aktiengesellschaft Method for starting a steam turbine plant
EP1934260B1 (en) 2005-10-14 2017-05-17 Innate Pharma Compositions and methods for treating proliferative disorders
AU2006323027B2 (en) 2005-12-05 2012-08-02 Pfizer Products Inc Method of treating abnormal cell growth
CN101326175B (en) 2005-12-05 2012-07-18 辉瑞产品公司 Polymorphs of a C-MET/HGFR inhibitor
WO2007113648A2 (en) 2006-04-05 2007-10-11 Pfizer Products Inc. Ctla4 antibody combination therapy
DK2526933T3 (en) 2006-09-22 2015-05-18 Pharmacyclics Inc Inhibitors of Bruton's tyrosine kinase
CN105037549B (en) 2007-01-11 2018-09-28 诺和诺德公司 Anti-KIR antibodies, preparation and its application
JP2008278814A (en) 2007-05-11 2008-11-20 Igaku Seibutsugaku Kenkyusho:Kk Release of immunoregulation by agonistic anti-human gitr antibody, and application thereof
PL2170959T3 (en) 2007-06-18 2014-03-31 Merck Sharp & Dohme Antibodies to human programmed death receptor pd-1
PT2175884T (en) 2007-07-12 2016-09-21 Gitr Inc Combination therapies employing gitr binding molecules
JP5535074B2 (en) 2007-10-01 2014-07-02 ブリストル−マイヤーズ スクウィブ カンパニー Human antibodies that bind to mesothelin and uses thereof
NZ586053A (en) 2007-11-09 2012-09-28 Peregrine Pharmaceuticals Inc Anti-vegf antibody compositions and methods
US20110085970A1 (en) 2007-11-30 2011-04-14 Terrett Jonathan A Anti-b7h4 monoclonal antibody-drug conjugate and methods of use
AR072999A1 (en) 2008-08-11 2010-10-06 Medarex Inc HUMAN ANTIBODIES THAT JOIN GEN 3 OF LYMPHOCYTARY ACTIVATION (LAG-3) AND THE USES OF THESE
WO2010065939A1 (en) 2008-12-05 2010-06-10 Indiana University Research & Technology Corporation Combination therapy to enhace nk cell mediated cytotoxicty
PL2376535T3 (en) 2008-12-09 2017-09-29 F.Hoffmann-La Roche Ag Anti-pd-l1 antibodies and their use to enhance t-cell function
KR101790802B1 (en) 2009-09-03 2017-10-27 머크 샤프 앤드 돔 코포레이션 Anti-gitr antibodies
WO2011130434A2 (en) 2010-04-13 2011-10-20 Celldex Therapeutics Inc. Antibodies that bind human cd27 and uses thereof
WO2011152351A1 (en) 2010-05-31 2011-12-08 小野薬品工業株式会社 Purinone derivative
WO2012071411A2 (en) 2010-11-22 2012-05-31 Innate Pharma Sa Nk cell modulating treatments and methods for treatment of hematological malignancies
WO2012122444A1 (en) 2011-03-10 2012-09-13 Provectus Pharmaceuticals, Inc. Combination of local and systemic immunomodulative therapies for enhanced treatment of cancer
SG195082A1 (en) 2011-05-25 2013-12-30 Innate Pharma Sa Anti-kir antibodies for the treatment of inflammatory disorders
WO2013017989A1 (en) 2011-08-02 2013-02-07 Pfizer Inc. Crizotinib for use in the treatment of cancer
WO2013039954A1 (en) 2011-09-14 2013-03-21 Sanofi Anti-gitr antibodies
EP2776032B1 (en) 2011-11-09 2018-10-17 Bristol-Myers Squibb Company Treatment of hematologic malignancies with an anti-cxcr4 antibody
US10864271B2 (en) * 2012-02-15 2020-12-15 Rutgers, The State University Of New Jersey Combination therapy using riluzole to enhance tumor sensitivity to ionizing radiation
US9856320B2 (en) 2012-05-15 2018-01-02 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting PD-1/PD-L1 signaling
KR101566539B1 (en) 2012-06-08 2015-11-05 국립암센터 Novel epitope for switching to Th2 cell and use thereof
UY34887A (en) 2012-07-02 2013-12-31 Bristol Myers Squibb Company Una Corporacion Del Estado De Delaware OPTIMIZATION OF ANTIBODIES THAT FIX THE LYMPHOCYTE ACTIVATION GEN 3 (LAG-3) AND ITS USES
PL2904011T3 (en) 2012-10-02 2018-01-31 Bristol Myers Squibb Co Combination of anti-kir antibodies and anti-pd-1 antibodies to treat cancer
AR097306A1 (en) 2013-08-20 2016-03-02 Merck Sharp & Dohme MODULATION OF TUMOR IMMUNITY
TW201605896A (en) 2013-08-30 2016-02-16 安美基股份有限公司 GITR antigen binding proteins
ES2728578T3 (en) 2013-09-20 2019-10-25 Bristol Myers Squibb Co Combination of anti-LAG-3 antibodies and anti-PD-1 antibodies to treat tumors
WO2015061752A1 (en) 2013-10-25 2015-04-30 Pharmacyclics, Inc. Treatment using bruton's tyrosine kinase inhibitors and immunotherapy
SG11201609721WA (en) 2014-05-28 2016-12-29 Agenus Inc Anti-gitr antibodies and methods of use thereof
CA2954868C (en) * 2014-07-11 2023-08-29 Genentech, Inc. Anti-pd-l1 antibodies and diagnostic uses thereof
US20160073759A1 (en) * 2014-09-15 2016-03-17 Beauty Solutions Agency Inc. Nail covering
ES2843324T3 (en) * 2014-11-18 2021-07-16 Maximum Fidelity Surgical Simulations Llc Reconstitution of the post-mortem circulation
US20160140879A1 (en) * 2014-11-19 2016-05-19 David Hananel Anatomically correct movement or deformation of simulated bodily structures
DK3265448T3 (en) * 2015-03-03 2022-03-28 Biohaven Therapeutics Ltd RILUZOLE PRODUCT AND ITS USE
US11911369B2 (en) * 2015-03-03 2024-02-27 Biohaven Therapeutics Ltd. Prodrugs of riluzole and their method of use
MY188749A (en) 2015-04-17 2021-12-28 Bristol Myers Squibb Co Compositions comprising a combination of nivolumab and ipilimumab
US20190054090A1 (en) 2015-10-01 2019-02-21 Gilead Sciences, Inc. Combination of a btk inhibitor and a checkpoint inhibitor for treating cancers
MX2018013868A (en) * 2016-05-20 2019-02-21 Biohaven Pharm Holding Co Ltd Use of glutamate modulating agents with immunotherapies to treat cancer.

Patent Citations (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5631023A (en) 1993-07-09 1997-05-20 R.P. Scherer Corporation Method for making freeze dried drug dosage forms
US6212791B1 (en) 1993-10-01 2001-04-10 R.P. Scherer Corporation Method of applying indicia to a fast-dissolving dosage form
US5837287A (en) 1994-10-28 1998-11-17 R P Scherer Corporation Process for preparing solid pharmaceutical dosage forms
US6471992B1 (en) 1997-02-20 2002-10-29 Therics, Inc. Dosage form exhibiting rapid disperse properties, methods of use and process for the manufacture of same
US6149938A (en) 1997-07-25 2000-11-21 Elan Pharma International Limited Process for the preparation of a granulate suitable to the preparation of rapidly disintegrable mouth-soluble tablets and compositions obtained thereby
US7939105B2 (en) 1998-11-20 2011-05-10 Jagotec Ag Process for preparing a rapidly dispersing solid drug dosage form
US6465010B1 (en) 1999-08-04 2002-10-15 Drugtech Corporation Means for creating a mass having structural integrity
US6284270B1 (en) 1999-08-04 2001-09-04 Drugtech Corporation Means for creating a mass having structural integrity
US6316029B1 (en) 2000-05-18 2001-11-13 Flak Pharma International, Ltd. Rapidly disintegrating solid oral dosage form
US6982251B2 (en) 2000-12-20 2006-01-03 Schering Corporation Substituted 2-azetidinones useful as hypocholesterolemic agents
US6814978B2 (en) 2000-12-29 2004-11-09 Mcneil-Ppc, Inc. Process for preparing a soft tablet
US6509040B1 (en) 2001-06-22 2003-01-21 R.P. Scherer Corporation Fast dispersing dosage forms essentially free of mammalian gelatin
US6908626B2 (en) 2001-10-12 2005-06-21 Elan Pharma International Ltd. Compositions having a combination of immediate release and controlled release characteristics
US7993674B2 (en) 2002-02-13 2011-08-09 Weibel Michael K Drug dose-form and method of manufacture
US7282217B1 (en) 2003-08-29 2007-10-16 Kv Pharmaceutical Company Rapidly disintegrable tablets
US7425341B1 (en) 2003-08-29 2008-09-16 K.V. Pharmaceutical Company Rapidly disintegrable tablets
US8158152B2 (en) 2005-11-18 2012-04-17 Scidose Llc Lyophilization process and products obtained thereby
US8048449B2 (en) 2005-12-27 2011-11-01 Jubilant Organosys Ltd. Mouth dissolving pharmaceutical composition and process for preparing the same
US8127516B2 (en) 2007-06-27 2012-03-06 Hanmi Pharm. Co., Ltd. Method for preparing rapidly disintegrating formulation for oral administration and apparatus for preparing and packing the same
US8221480B2 (en) 2008-10-31 2012-07-17 The Invention Science Fund I, Llc Compositions and methods for biological remodeling with frozen particle compositions
US8256233B2 (en) 2008-10-31 2012-09-04 The Invention Science Fund I, Llc Systems, devices, and methods for making or administering frozen particles
US8313768B2 (en) 2009-09-24 2012-11-20 Mcneil-Ppc, Inc. Manufacture of tablet having immediate release region and sustained release region
US20160019787A1 (en) 2011-10-20 2016-01-21 Apple Inc. Method for Locating a Vehicle
WO2013138753A1 (en) 2012-03-16 2013-09-19 Fox Chase Chemical Diversity Center, Inc. Prodrugs of riluzole and their method of use
US20150045401A1 (en) * 2012-03-16 2015-02-12 Fox Chase Chemical Diversity Center, Inc. Prodrugs of riluzole and their method of use
US20150148329A1 (en) * 2012-06-23 2015-05-28 Fox Chase Chemical Diversity Center, Inc. Pro-drugs of riluzole and their method of use for the treatment of amyotrophic lateral sclerosis
WO2016073759A1 (en) * 2014-11-05 2016-05-12 The Regents Of The University Of California Combination immunotherapy

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
HIRANO FKANEKO KTAMURA H ET AL.: "Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity.", CANCER RES., vol. 65, 2005, pages 1089 - 1096, XP002419626
PREUSSER, M ET AL.: "Prospects of immune checkpoint modulators in the treatment of glioblastoma", NATURE REVIEWS NEUROLOGY, vol. 11, no. Issue 9, September 2015 (2015-09-01), pages 1 - 22, XP055438601 *
VELICU SULASOV IV ET AL.: "A safety and efficacy study of local delivery of interleukinl2 transgene by PPC polymer in a model of experimental glioma", ANTICANCER DRUGS, vol. 19, 2008, pages 133 - 142
ZENG, J. ET AL., INT J RADIAT ONCOL BIOL PHYS, vol. 86, no. 2, 1 June 2013 (2013-06-01), pages 343 - 349

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210236471A1 (en) * 2015-03-03 2021-08-05 Biohaven Therapeutics Ltd. Riluzole prodrugs and their use
US11168144B2 (en) 2017-06-01 2021-11-09 Cytomx Therapeutics, Inc. Activatable anti-PDL1 antibodies, and methods of use thereof
WO2019164861A1 (en) * 2018-02-21 2019-08-29 AI Therapeutics, Inc. Combination therapy with apilimod and glutamatergic agents
US10751345B2 (en) 2018-02-21 2020-08-25 AI Therapeutics, Inc. Combination therapy with apilimod and glutamatergic agents
CN111801098A (en) * 2018-02-21 2020-10-20 人工智能治疗公司 Combination therapy with apilimod and glutamatergic agents
US11439649B2 (en) 2018-02-21 2022-09-13 AI Therapeutics, Inc. Combination therapy with apilimod and glutamatergic agents
CN114222729A (en) * 2019-06-12 2022-03-22 范德比尔特大学 Amino acid transport inhibitors and uses thereof
WO2023202652A1 (en) * 2022-04-21 2023-10-26 Jacobio Pharmaceuticals Co., Ltd. Pharmaceutical combination and use thereof
US11957688B2 (en) 2022-07-12 2024-04-16 OrphAl Therapeutics Inc. Combination therapy with apilimod and glutamatergic agents

Also Published As

Publication number Publication date
EP3458053B1 (en) 2021-12-08
PT3458053T (en) 2022-02-01
LT3458053T (en) 2022-02-25
EP4019019A1 (en) 2022-06-29
MX2022010377A (en) 2022-09-21
KR102369735B1 (en) 2022-03-02
CA3025019A1 (en) 2017-11-23
RS62935B1 (en) 2022-03-31
JP2019516712A (en) 2019-06-20
DK3458053T3 (en) 2022-02-21
CY1124999T1 (en) 2023-01-05
KR102525527B1 (en) 2023-04-24
PH12018502409A1 (en) 2019-04-08
MX2018013868A (en) 2019-02-21
SI3458053T1 (en) 2022-04-29
IL292302A (en) 2022-06-01
JP2023012508A (en) 2023-01-25
IL292302B2 (en) 2023-10-01
AU2017266951A1 (en) 2018-11-22
WO2017201502A1 (en) 2017-11-23
JP7169195B2 (en) 2022-11-10
CN115350263A (en) 2022-11-18
AU2021221560B2 (en) 2023-10-26
IL262849B (en) 2022-05-01
IL262849A (en) 2018-12-31
IL292302B1 (en) 2023-06-01
ES2905823T3 (en) 2022-04-12
JP2019516711A (en) 2019-06-20
CN109890387A (en) 2019-06-14
HUE058184T2 (en) 2022-07-28
EP3458053A1 (en) 2019-03-27
KR20190009334A (en) 2019-01-28
JP7224186B2 (en) 2023-02-17
BR112018073781A2 (en) 2019-02-26
EP3458053A4 (en) 2019-11-20
EP3458054A1 (en) 2019-03-27
EA201892587A1 (en) 2019-04-30
US11400155B2 (en) 2022-08-02
US20190175731A1 (en) 2019-06-13
AU2017266951B2 (en) 2021-05-27
ES2912131T3 (en) 2022-05-24
EP4019019B1 (en) 2024-02-07
EP3458054A4 (en) 2019-10-02
AU2021221560A1 (en) 2021-09-16
KR20220029772A (en) 2022-03-08
US20190290619A1 (en) 2019-09-26
JP2022191256A (en) 2022-12-27
ZA201807398B (en) 2020-05-27
PL3458053T3 (en) 2022-04-25
EP3458054B1 (en) 2022-02-23
SG11201809669RA (en) 2018-11-29
CN109890387B (en) 2022-06-14
HRP20220237T1 (en) 2022-04-29
US20230310595A1 (en) 2023-10-05

Similar Documents

Publication Publication Date Title
US20230310595A1 (en) Use of riluzole, riluzole prodrugs or riluzole analogs with immunotherapies to treat cancers
TWI767976B (en) Methods of treating cancer with anti-pd-1 antibodies
JP2021183650A (en) Therapeutic compositions, combinations, and methods of use
US11622961B2 (en) Combination therapies for treating cancer
CN113395967A (en) Pharmaceutical combination comprising TNO155 and PD-1 inhibitor
CN108136025A (en) A kind of novel method using immune modulating treatment cancer
JP2018507220A (en) Methods, compositions and kits for the treatment of cancer
JP6753860B2 (en) Sublingual preparation of riluzole
JP7388635B2 (en) Biomarkers to determine the effectiveness of immune checkpoint inhibitors
CN111132696B (en) Use of PD-1 antibodies in combination with epigenetic modulators for the preparation of a medicament for the treatment of tumors
CN112292128A (en) EP4 inhibitors and uses thereof
TWI747868B (en) Antitumor agent containing immunomodulator and antitumor effect potentiator
WO2018072743A1 (en) Use of pd-1 antibody conjugated with ido inhibitor in preparing anti-tumor drug
WO2017205216A1 (en) Combination of pembrolizumab and abemaciclib for the treatment of cancer
TW202038942A (en) Combination therapies for treating disease using an innate immunity modifier and an ox40 agonist
CN109663130B (en) Use of a combination of a PD-1 antibody and a MEK inhibitor for the preparation of a medicament for the treatment of tumors
JP2022500485A (en) Grapiplant unit dosage form
WO2020119757A1 (en) Use of cdk4/6 inhibitor in combination with immunotherapy in preparing drug for treating lymphoma
CN117222663A (en) Methods of treating cancer using multi-specific binding proteins that bind NKG2D, CD16 and tumor-associated antigens
EP4342492A1 (en) Pharmaceutical combination and use thereof
CN112439060B (en) New use of PD-L1 immunotherapy
TW202346342A (en) Drug combination of anti-TIM-3 antibody and anti-PD-1 antibody
NZ788180A (en) Combination therapy with notch and pd-1 or pd-l1 inhibitors

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 2018560520

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17800310

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2017800310

Country of ref document: EP

Effective date: 20181220